MXPA06004843A - Compositions and methods for detecting and treating diseases and conditions related to chemokine receptors - Google Patents

Compositions and methods for detecting and treating diseases and conditions related to chemokine receptors

Info

Publication number
MXPA06004843A
MXPA06004843A MXPA/A/2006/004843A MXPA06004843A MXPA06004843A MX PA06004843 A MXPA06004843 A MX PA06004843A MX PA06004843 A MXPA06004843 A MX PA06004843A MX PA06004843 A MXPA06004843 A MX PA06004843A
Authority
MX
Mexico
Prior art keywords
ccx
ckr2
cancer
agent
cell
Prior art date
Application number
MXPA/A/2006/004843A
Other languages
Spanish (es)
Inventor
M Burns Jennifer
Summers Bretton
C Howard Maureen
J Schall Thomas
Miao Zhenhua
Original Assignee
Chemocentryx Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Chemocentryx Inc filed Critical Chemocentryx Inc
Publication of MXPA06004843A publication Critical patent/MXPA06004843A/en

Links

Abstract

Ligands of CCX-CKR2 and the biological role of CCX-CKR2 in cancer is described.

Description

COMPOSITIONS AND METHODS FOR DETECTING AND TREATING DISEASES AND CONDITIONS RELATED TO BURNER RECEPTORS BACKGROUND OF THE INVENTION Chemokines constitute a family of small cytokines that occur in inflammation and regulate the recruitment, activation and proliferation of leukocytes (Baggiolini, M. et al., Adv. Immunol. 97-179 (1994); Springer, T. A., Annu Rev. Physiol. 57: 827-872 (nineteen ninety five); and Schalí, T. J. and K. B. Bacon, Curr. Opin. Immunol. 6: 865-873 (1994)). Chemokines are capable of selectively inducing chemotaxis of formed elements of blood (other than red blood cells), including leukocytes such as neutrophils, monocytes, macrophages, eosinophils, basophils, mast cells and lymphocytes, including T cells and B cells. In addition to stimulating chemotaxis, other changes may be selectively induced by chemokines in responsive cells, including changes in cell shape, transient elevations of intracellular free calcium ion concentration (Ca2 +), granule exocytosis, up-regulation of integrin, formation of bioactive lipids (for example, leukotrienes) and respiratory arrest, associated with leukocyte activation. Thus, chemokines are early activators of the inflammatory response, causing inflammatory mediator release, chemotaxis and extravasation to sites of infection or inflammation. Two chemokine subfamilies, designated chemokines CXC and CC, are distinguished by the arrangement of the first two of the four conserved cysteine residues, which are either separated by an amino acid (as in chemokines CXC SDF-1, 1L-8, IP-10, MIG, PF4, ENA-78, GCP-2, GRO-a, GRO-β, GRO- ?, NAP-2, NAP-4, I-TAC) or are adjacent residues (as in CC chemokines) , MlP-la, MlP-lβ, RANTES, MCP-1, MCP-2, MCP-3, 1-309). Most CXC chemokines attract neutrophil leukocytes. For example, the chemokines CXC interieucine 8 (IL-8), platelet factor 4 (PF4) and neutrophil activation peptide 2 (NAP-2) are chemoattractants and potent activators of neutrophils. The CXC chemokines designated MIG (gamma interferon-induced onoquine) and IP-10 (interferon-α-induced 10 kDa protein) are particularly active in inducing the chemotaxis of activated peripheral blood lymphocytes. The CC chemokines are generally less selective and can attract a variety of leukocyte cell types, including monocytes, eosinophils, basophils, T lymphocytes and natural killer cells. CC chemokines such as human monocyte chemotactic proteins 1-3 (MCP-1, MCP-2 and MCP-3), RANTES (Regulated in Activation, Normal T Expressed and Secreted), and macrophage inflammatory proteins and lß (MlP-la and MIP-Iß) have been characterized as chemoattractants and activators of monocytes or lymphocytes, but they do not appear to be chemoattractants for neutrophils. The chemokines CC and CXC act through receptors belonging to a superfamily of seven G protein-coupled receptors spanning the transmembrane (Murphy, P.M., Pharmacol Rev. 52: 145-176 (2000)). This family of G protein coupled receptors comprises a large group of integral membrane proteins, which contain seven regions encompassing the transmembrane. The receptors are coupled to G proteins, which are heterotrimeric regulatory proteins capable of binding GTP and mediating signal transduction of coupled receptors, for example, by the production of intracellular mediators. Generally speaking, the chemokine receptor and chemokine interactions tend to be promiscuous in that a chemokine can bind many chemokine receptors and conversely a single chemokine receptor can interact with several chemokines. There are a few exceptions to this rule; One such exception has been the interaction between SDF-1 and CXCR4 (Bleul et al., J Exp Med, 184 (3): 1101-9 (1996); Oberlin et al., Nature, 382 (6594): 833-5 (1996). Originally identified as a B cell pre-growth stimulation factor (Nagasawa et al., Proc Acad Sci E U A, 91 (6): 2305-9 (1994)), SDF-1 has been the only human ligand reported for CXCR4. The SDF-1 gene encodes two proteins, designated SDF-la and SDF-1, by alternative splicing. These two proteins are identical except for the four amino acid residues that are present in the carboxyl-terminal SDF-lβ and absent from SDF-la. There are many aspects of chemokine receptor signaling and selectivity for ligands that were not previously understood. For example, there are a number of orphan receivers for which the function has not been previously determined. RDC1, for example, although earlier it is thought to be a receptor for vasoactive intestinal peptide (VIP), is now considered to be an orphan receptor because its endogenous ligand has not been identified. See, for example, Cook et al. (2): 149-152 (1992). The present invention addresses these and other points. BRIEF DESCRIPTION OF THE INVENTION The present invention provides methods for identifying an agent that binds CCX-CKR2 on a cell. In some embodiments, the method comprises contacting a plurality of agents with a CCX-CKR2 polypeptide comprising an extracellular domain at least 95% identical to an extracellular domain of SEQ ID NO: 2, or a fragment thereof which binds SDF1 or I-TAC; and selecting an agent that competes with I-TAC or SDF1 to bind to the CCX-CKR2 polypeptide or fragment thereof, in order to thereby identify an agent that binds CCX-CKR2 on a cell. In some modalities, the cell is a cancer cell. In some embodiments, the method further comprises testing the agent selected for the ability to bind to, or inhibit the growth of, a cell. In some modalities, the cell is a cancer cell. In some embodiments, the method further comprises testing the agent selected for the ability to alter kidney function. In some embodiments, the method further comprises testing the agent selected for the ability to alter the brain or neuronal function. In some embodiments, the method further comprises testing the agent selected for the ability to change cell adhesion to endothelial cells. In some modalities, the agent is less than 1,500 daltons. In some embodiments, the agent is an antibody. In some embodiments, the agent is a polypeptide. In some embodiments, the CCX-CKR2 polypeptide comprises the sequence shown in SEQ ID NO: 2. The present invention also provides methods for determining the presence or absence of a cancer cell. In some embodiments, the method comprises contacting a sample comprising a cell with an agent that specifically binds to SEQ ID NO: 2, and detecting the binding of the agent to a polypeptide in the sample, wherein the binding of the agent to the the sample indicates the presence of a cancer cell. In some embodiments, the agent is an antibody. In some modalities, the agent is less than 1,500 daltons. In some embodiments, the agent is a polypeptide. In some embodiments, the polypeptide detected is SEQ ID NO: 2. In some embodiments, the sample is from a human. In some modalities, the method is used to diagnose cancer in a human. In some modalities, the method is used to provide a prognosis of cancer in a human. In some embodiments, the cancer is selected from the group consisting of cervical cancer, breast cancer, lymphoma, glioblastomas, prostate cancer and leukemia. In some embodiments, the cancer is not Kaposi's sarcoma, multicentric Castleman's disease or primary fusion lymphoma associated with AIDS. In some embodiments, the antibody competes with SDF1 and I-TAC for binding to SEQ ID NO: 2. The present invention also provides methods for providing a diagnosis or prognosis of an individual having cancer. In some modalities, the method comprises detecting the presence or absence of the expression of a polynucleotide encoding a CCX-CKR2 polypeptide in a cell of an individual, wherein the CCX-CKR2 polypeptide binds SDF1 and / or SDF-1 and the CCX-CKR2 polypeptide it is at least 95% identical to SEQ ID NO: 2, in order to diagnose a cancer in the individual. In some embodiments, the CCX-CKR2 polypeptide is shown in SEQ ID NO: 2. In some embodiments, the cancer is selected from the group consisting of cervical cancer, breast cancer, lymphoma, glioblastomas, prostate cancer and leukemia. In some embodiments, the cancer is not Kaposi's sarcoma, multicentric Castleman's disease or primary fusion lymphoma associated with AIDS. The present invention also provides antibodies that specifically compete with SDF-1 and I-TAC for binding to SEQ ID NO: 2. In some embodiments, the antibody is a monoclonal antibody. In some embodiments, the antibody is a humanized antibody. The present invention also provides methods comprising contacting a cell with an agent that specifically binds to SEQ ID NO: 2, wherein the agent competes with SDF-1 and I-TAC for binding to a CCX-CKR2 polypeptide, and wherein the cell expresses a CCX-CKR2 polypeptide comprising an extracellular domain at least 95% identical to an extracellular domain of SEQ ID NO: 2, in order to bind the agent to the CCX-CKR2 polypeptide on the cell. In some modalities, the agent is less than 1,500 daltons. In some embodiments, the agent is an antibody. In some embodiments, the agent is a polypeptide. In some embodiments, the CCX-CKR2 polypeptide is as shown in SEQ ID NO: 2. In some embodiments, the agent is identified by a method comprising contacting a plurality of agents with a CCX-CKR2 polypeptide comprising a extracellular domain at least 95% identical to an extracellular domain of SEQ ID NO: 2, or a fragment thereof that binds SDFl or 1-TAC; and selecting an agent that competes with I-TAC or SDF-1 for binding to the CCX-CKR2 polypeptide or fragment thereof, in order to thereby identify an agent that binds to a cancer cell. The present invention also provides methods for treating cancer in an individual. In some embodiments, the methods comprise administering to the individual a therapeutically effective amount of a polynucleotide that inhibits the expression of a CCX-CKR2 polynucleotide. In some embodiments, the polynucleotide CCX-CKR2 encodes SEQ ID NO: 2. In some embodiments, the polynucleotide CCX-CKR2 comprises SEQ ID NO: 1. In some embodiments, the polynucleotide administered inhibits expression via a siRNA. The present invention also provides methods for treating cancer in an individual. In some embodiments, the method comprises administering to the individual a therapeutically effective amount of an agent competing with SDF1 and I-TAC for binding to SEQ ID NO: 2. In some embodiments, the agent is less than 1,500 daltons. In some embodiments, the agent is an antibody. In some embodiments, the agent is a polypeptide. In some embodiments, the agent is identified by a method comprising contacting a plurality of agents with a CCX-CKR2 polypeptide comprising an extracellular domain at least 95% identical to an extracellular domain of SEQ ID NO: 2 or a fragment of the same that links SDFl or I-TAC; and selecting an agent that competes with I-TAC or SDF-1 for binding to the CCX-CKR2 polypeptide or fragment thereof, in order to thereby identify an agent that binds a cancer cell. In some embodiments, the cancer is selected from the group consisting of cervical cancer, breast cancer, lymphoma, glioblastomas, prostate cancer and leukemia. In some modalities, the cancer is not Kaposi's sarcoma, multicentric Castleman's disease or primary fusion lymphoma associated with AIDS. BRIEF DESCRIPTION OF THE DRAWINGS Figures 1A-I illustrate the link data demonstrating a different SDF-1 finger link printing on different cell types. The link competition profile .using 125ISDF-la with the radioligand probe in (a) CEM-NKr (Figures 1A-C) and (b) MCF-7 (Figures 1D-F), as well as 125I I-TAC ( Figures 1G-I) used as the radioligand probe in (c) MCF-7, in a binding site experiment with a comprehensive array of > 90 chemokines and variants of discrete viral, human and murine chemokines as cold competitors. The percent inhibition of radioligand binding is shown as a bar graph and reveals that SDF-la and I-TAC are cross-displaced on MCF-7 but not on CEM-NKr cells. White bars, high potential affinity (inhibition> 80%); gray bars, moderate at low potential affinity (inhibition between 60-79%, black bars, little or no affinity (inhibition <60%) .The results are the average of three determinations.The error bars are omitted for clarity. Figure 2 illustrates a comparison of ligand binding affinity and specificity on CEN-NKr and MCF-7 The selected, high potential affinity ligands identified in Figure 2 were selected for dose response competition on CEN- NKr (empty frames) and MCF-7 (full frames) In each competition 125I SDF-la is in competition with a cold competing chemokine as indicated.Figure 3 illustrates the 125I I-TAC binding on MCF-7 cells it is not due to a classical CXCR3 binding interaction.The ability of 125I-TAC to compete with the chemokines indicated was examined in the presence of regulatory solution only (filled squares), excess MIG (to inhibit any medium binding). Adopted by CXCR3; empty triangles), or in excess SDF-la (asterisk). Figure 4 illustrates that the binding phenotype described herein can be recapitulated in a cell line that does not endogenously express this receptor. The MDA-MB 435s line of breast tumor stably transfected with CCX-CKR2 exhibits the 125I-SDF-la binding. This link can be competed with the SDF-la and I-TAC cold. By comparison wild-type (non-transfected) cells do not give a binding signal of 1251 SDF-the productive one. Figure 5 illustrates the competitive link data. Two small molecules, CCX0803 (full circles) and CCX7923 (empty circles), compete specifically with the 125I SDF-la, on discrete cell types; no cross competition is detected. SDF-sDF-la (asterisk) was also included as a cold competitor of the 125I SDF-la link on both MCF-7 and CEM-NKr. The chemical structures of CCX0803 and CCX7923 are shown in the array. The predicted IC5o values of SDF-la and antagonist competition of CXCR4 are given in the accompanying table. Figure 6 illustrates the effect of treatment of mammary carcinoma cells, expressing CCX-CKR2, with a small molecule CCX-CKR2 antagonist compared to cells not treated with the antagonist. Figure 7 illustrates that antagonist 3451 of CCX-CKR2 (see, Table 1, Compound No. 49) inhibits the adhesion of cells expressing CCX-CKR2 to a vascular endothelial monolayer. Figure 8 illustrates that antagonism of CCX-CKR2 on mammary carcinoma cells reduces tumor volume. DEFINITIONS "Chemokines" or "chemokine ligand" refers to a small protein composed of approximately 50 to 110 amino acids and which shares sequence homology with other known chemokines (see, for example, Murphy, PM, Pharmacol Rev. 52: 145- 176 (2000)). The chemokines are classified according to the relative positions of the first pair of cysteines (Cs) found in the primary amino acid sequence. In the CXCL chemokines, the first pair of cysteines is separated by any individual amino acid. The CCL chemokines have adjacent cysteines. In the CX3CL chemokines, the first pair of cysteines is separated by 3 amino acids. The CL chemokines contain only a single cysteine in the homologous position. Chemokines can activate biological function by binding to and activating chemokine receptors. A "chemokine receptor" refers to a polypeptide that specifically interacts with a chemokine molecule. A chemokine receptor is typically a G protein-coupled receptor with seven transmembrane domains. The chemokine receptors may possess several common structural features including a highly acidic N-terminal domain; the DRYLAIVHA amino acid sequence (or a minor variation of that sequence) found within the second extracellular circuit of many chemokine receptors; a third short intracellular circuit with a total basic load; a cysteine residue with each of the four extracellular domains. Typically, chemokine receptors have a total size of approximately 340-370 amino acid residues. See, for example, reviewed in Murphy, P. M. Chemokine Receptors; Overview, Academia Press 2000; Oy Loetscher P. and Clark-Lewis I. J. Leukoci te Biol. 69: 881 (2001). Exemplary chemokine receptors include, for example, CC-chemokine receptors (including CCR1, CCR2, CCR3, CCR4, CCR5, CCR6, CCR7, CCR8, CCR9 and CCR10), CXC-chemokine receptors (including CXCR1, CXCR2, CXCR3, CXCR4, CXCR5, CXCR6 and CCX-CKR2 (eg, SEQ ID N0: 2)), CX3CR1, CXR1, CCXCKR (CCR11), virally encoded chemokine receptors, US28, ECRF3, Herpesvirus GPCR associated with Kaposi's sarcoma ( 0RF74), receptors coupled to the G protein bound to the Poxvirus membrane; D6 and DARC. Defined responses that can be stimulated by chemokine receptors include transmembrane signaling, activation of cytoplasmic signaling cascades, cytoskeletal rearrangement, adhesion, chemotaxis, invasion, metastasis, cytokine production, gene induction, gene repression, induction of protein expression, or modulation of cell growth and differentiation, including the development of cancer. "RDC1", herein referred to as "CCX-CKR2" refers to a G-protein coupled receptor budget of seven transmembrane domain (GPCR). The dog orthologue CCX-CKR2 was originally identified in 1991. See, Libert et al., Science 244: 569-572 (1989). The sequence of the dog is described in Libert et al., Nuc. Acids Res. 18 (7): 1917 (1990). The mouse sequence is described in, for example, Essen et al., Iiamunogenetics 47: 364-370 (1998). The human sequence is described in, for example, Sreedharan et al., Proc. Nati Acad. Sci. USA 88: 4986-4990 (1991) erroneously described the protein as a vasoactive intestinal peptide receptor. "CCX-CKR2" includes sequences that are substantially similar to conservatively modified variants of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO : 7, SEQ ID NO: 8, SEQ ID NO: 9 or SEQ ID NO: 10. The terms "peptide mimetic" and "mimetic" refer to a synthetic chemical compound having substantially the same structural and functional characteristics as antagonists or agonists. of a chemokine receptor. Peptide analogs are commonly used in the pharmaceutical industry as non-peptide drugs with analogous properties to those of the template peptide. These types of non-peptide compounds are called "peptide mimetics" or "peptidomimetics" (Fauchere, J. Adv. Drug Res. 15:29 (1986)).; Veber and Freidinger TINS p. 392 (1985); and Evans et al., J. Med. Chem. 30: 1229 (1987), which are incorporated herein by reference). Peptide mimetics that are structurally similar to therapeutically useful peptides can be used to produce an equivalent or increased therapeutic or prophylactic effect. Generally, peptidomimetics are structurally similar to a paradigm polypeptide (i.e., a polypeptide having a biological or pharmacological activity), but have one or more peptide bonds optionally replaced by a link selected from the group consisting of, for example , -CH2NH-, -CH2S-, -CH2-CH2-, -CH = CH- (cis and trans), -C0CH2-, -CH (0H) CH2- and CH2S0-. The mimetic may be either entirely composed of unnatural, synthetic analogs of amino acids, or, it is a chimeric molecule of partially natural peptide amino acids and non-natural analogs of amino acids. The mimetic can also incorporate any number of natural conservative amino acid substitutions as long as such substitutions do not substantially alter the structure of the mimetic and / or activity. A mimetic can mimic, for example, the linkage of SDF-1 or I-TAC or CCX-CKR2. For example, a mimetic composition that is within the scope of the invention if it is capable of inhibiting or increasing the function activity of CCX-CKR2. "Antibody" refers to a polypeptide substantially encoded by an immunoglobulin gene or immunoglobulin genes, or fragments thereof, that specifically bind and recognize an analyte (antigen). The recognized immunoglobulin genes include the kappa, lambda, alpha, gamma, delta, epsilon and mu constant region genes, as well as a number of immunoglobulin variable region genes. Light chains are classified as either kappa or lambda. The heavy chains are classified as gamma, mu, alpha, delta or epsilon, which in turn define the classes of immunoglobulin, IgGel IgM, IgA, IgD and IgE respectively. An exemplary immunoglobulin structural unit (antibody) comprises a tetramer. Each tetramer is composed of two identical pairs of polypeptide chains, each pair having a "light" chain (approximately 25 kD) and a "heavy" chain (approximately 50-70 kD). The N-terminal of each chain defines a variable region of approximately 100 to 110 or more amino acids mainly responsible for the recognition of antigens. The terms variable light chain (VL) and variable heavy chain (VH) refer to these light and heavy chains, respectively. Antibodies exist, for example, as intact immunoglobulins or as a number of well-characterized fragments produced by digestion with various peptidases. Thus, for example, pepsin digests an antibody below disulfide bonds in the hindered region to produce F (ab) '2, a Fab dimer which by itself is a light chain linked to VH-CH1 via a bond of disulfide. The F (ab) '2 can be reduced under moderate conditions to break the disulfide bond in the hindered region, thereby converting the F (ab) A dimer into a Fab' monomer. The Fab 'monomer is essentially an Fab with part of the hindered region (see, Paul (Ed.) Fundamental Immunology, Third Edition, Raven Press, NY (1993)). While several antibody fragments are defined in terms of the digestion of an intact antibody, one skilled in the art will appreciate that such fragments can be synthesized de novo either chemically or by using the recombinant DNA methodology. Thus, the term "antibody" as used herein, also includes antibody fragments either produced by the modification of whole antibodies or those synthesized de novo using recombinant DNA methodologies (e.g., single chain Fv). "Humanized" antibodies refers to a molecule having an antigen binding site that is substantially derived from an immunoglobulin of a non-human species and the remaining immunoglobulin structure of the molecule based on the structure and / or sequence of a human immunoglobulin. The antigen binding site can comprise either complete variable domains fused over constant domains or only regions of complementarity determination (CDRs) grafted onto regions of appropriate structure in the variable domains. The antigen binding sites may be wild-type or modified by one or more amino acid substitutions, for example, modified to resemble human immunoglobulin more closely. Some forms of humanized antibodies retain all CDR sequences (eg, a humanized mouse antibody containing all six CDRs of mouse antibodies). Other forms of humanized antibodies have one or more CDRs (one, two, three, four, five, six) that are altered with respect to the original antibody. The phrase "specifically (or selectively) binds an antibody" or "specifically (or selectively) immunoreactive with", when referring to a protein or peptide, refers to a binding reaction that is determinative of the presence of the protein in the presence of a heterogeneous population of proteins or other biological substances. Thus, under the designated immunoassay conditions, the specified antibodies bind a particular protein and do not bind a significant amount of other proteins present in the sample. Specific binding to an antibody under such conditions may require that an antibody be selected for its specificity for a particular protein. For example, antibodies raised against a protein having an amino acid sequence encoded by any of the polynucleotides of the invention can be selected to obtain antibodies specifically immunoreactive with that protein and not with other proteins, except for polymorphic variants, eg, proteins. at least 80%, 85%, 90%, 95% or 99% identical to SEQ ID NO: 2. A variety of immunoassay formats can be used to select antibodies specifically immunoreactive with a particular protein. For example, solid-phase ELISA immunoassays, Western blot or immunohistochemistry are routinely used to select monoclonal antibodies specifically immunoreactive with a protein. See, Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Publications, NY (1988) for a description of immunoassay formats and conditions that can be used to determine specific immunoreactivity, typically, a specific or selective reaction will be so less twice the signal for background noise and more typically more than 10 to 100 times the background. A "ligand" refers to an agent, e.g., a polypeptide or other molecule, capable of binding to a chemokine receptor. As used herein, "an agent that binds to a chemokine receptor" refers to an agent that binds the chemokine receptor with high affinity. "High affinity" refers to a sufficient affinity to induce a pharmacologically relevant response, for example, the ability to compete significantly for binding to a natural chemokine ligand to a chemokine receptor at pharmaceutically relevant concentrations (e.g., at lower concentrations that approximately 10-5 M). See, for example, Example 1 and Figure 5. Some exemplary agents with high affinity will bind a chemokine receptor with an affinity greater than 10"6 M and sometimes greater than 107 M or 10" 8 M. An agent that fails to compete with the binding to a natural receptor ligand when the agent is in concentrations less than 10"14 M will be considered to be" binding "for the purposes of the invention The term" nucleic acid "or" polynucleotide "refers to deoxyribonucleotides or ribonucleotides and polymers thereof in the form of either a single or a double strand, unless specifically limited, the term comprises nucleic acids that contain known analogs of natural nucleotides that have similar binding properties as the reference nucleic acid and are metabolized in a manner similar to naturally occurring nucleotides, unless otherwise indicated, a particular nucleic acid sequence also implied citedly includes conservatively modified variants thereof (eg, degenerate codon substitutions) and complementary sequences as well as the explicitly indicated sequences. Specifically, degenerate codon substitutions can be achieved by generating sequences in which the third position of one or more selected (or all) codons is substituted with mixed base and / or deoxyinosin residues (Batzer et al., Nucleic Acid Res. 5081 (1991); Ohtsuka et al., J. Biol. Chem. 260: 2605-2608 (1985); and Cassol et al., (1992); Rossolini et al., Mol. Cell. Probes 8: 91-98 (1994)). The term nucleic acid is used interchangeably with the gene, cDNA and mRNA encoded by a gene. The terms "polypeptide", "peptide" and "protein" are used interchangeably herein to refer to a polymer of amino acid residues.The terms apply to amino acid polymers in which one or more amino acid residues is a mimetic artificial chemical of a naturally occurring corresponding amino acid, as well as polymers of naturally occurring amino acids and non-naturally occurring polymers of amino acids As used herein the term comprises chains of amino acids of any length, including full length proteins), in saying antigens) where the amino acid residues are linked by covalent peptide bonds The term "amino acid" refers to naturally occurring and synthetic amino acids, as well as amino acid analogs and amino acid mimetics that function in a manner similar to amino acids that occur naturally, the amino acids that occur n aturally are those encoded by the genetic code, as well as those amino acids that are subsequently modified, for example, hydroxyproline,? -carboxyglutamate and O-phosphoserine. The amino acid analogs refer to compounds having the same basic chemical structure as a naturally occurring amino acid, ie, a carbon a which is linked to a hydrogen, a carboxyl group, an amino group and a R group, by example, homoserin, norleucine, methionine sulphoxide methionine methyl sulfonium. Such analogs have modified R groups (e.g., norleucine) or modified peptide backbones, but retain the same basic chemical structure as a naturally occurring amino acid. "Amino acid mimetics" refers to chemical compounds that have structure that is different from the general chemical structure of an amino acid, but that functions in a manner similar to an amino acid that occurs naturally. The amino acids can be referred to herein by either their commonly known three letter symbols or by the letter symbols recommended by IPUAC-IUB Biochemical Commission Nomenclature. Nucleotides in the same way, can be referred by their commonly accepted single-letter codes. "Conservatively modified variants" apply to both amino acid and nucleic acid sequences. With respect to particular nucleic acid sequences, "conservatively modified variants" refers to those nucleic acids that encode identical or essentially identical amino acid sequences, or where the nucleic acid does not encode an amino acid sequence, to essentially identical sequences. Due to the degeneracy of the genetic code, a number of nucleic acid sequences will encode any given protein. For example, the GCA, GCC, GCG and GCU codons will all encode the amino acid alanine. Thus, in each position where an alanine is specified by a codon, the codon can be altered to any of the corresponding codons described without altering the encoded polypeptide. Such variations of nucleic acid are "silent variations" that are a kind of conservatively modified variations. Each nucleic acid sequence herein encodes a polypeptide also describes each possible silent variation of the nucleic acid. One skilled artisan will recognize that each codon in a nucleic acid (except AUG, which is ordinarily the only codon for methionine and TGG, which is ordinarily the only codon for tryptophan) can be modified to produce a functionally identical molecule. Accordingly, each silent variation of a nucleic acid encoding a polypeptide is implicit in each described sequence. As for amino acid sequences, an expert will recognize that individual substitutions, deletions or additions to a nucleic acid, peptide, polypeptide or protein sequence that alters, adds or deletes a single amino acid or a small percentage of amino acids in the encoded sequence is a "conservatively modified variant" where the alteration results in the substitution of an amino acid with a chemically similar amino acid. Conservative substitution tables that provide functionally similar amino acids are well known in the art. Such conservatively modified variants are in addition to and do not exclude the polymorphic variants, interspecies homologs and alleles of the invention. The following eight groups each contain amino acids that are conservative substitutions with each other: 1) Alanine (A), Glycine (G); 2) Aspartic acid (D), glutamic acid (E); 3) Asparagine (N), Glutamine (Q); 4) Arginine (R), Lysine (K); 5) Isoleucine (I), Leucine (L), Methionine (M), Valine (V); 6) Phenylalanine (F), Tyrosine (Y), Tryptophan (W); 7) Serine (S), threonine (T); and 8) Cysteine (C), Methionine (M) (see, for example, Creighton, Proteins (1984)). "Percentage of sequence identity" is determined by comparing two optimally aligned sequences on a comparison window, wherein the portion of the polynucleotide sequence in the comparison window may comprise additions or deletions (ie, space) as compared to the reference sequence (which does not include additions or deletions) for the optimal alignment of the two sequences. The percentage is calculated by determining the number of positions at which the identical nucleic acid base or amino acid residue occurs in both sequences to produce the number of equalized positions, by dividing the number of equalized positions by the total number of positions in the comparison window and multiply the result by 100 to produce the percentage of the sequence identity. The terms "identical" or "identity" percent in the context of two or more nucleic acids or polypeptide sequences, refers to two or more sequences or subsequences that are the same or have the specified percentage of amino acid or nucleotide residues. which are the same (i.e., 60% identity, optionally 65%, 70%, 75%, 80%, 85%, 90% or 95% identity over a specified region, eg, of the complete polypeptide sequences of the invention or the extracellular domains of the polypeptides of the invention), when compared and aligned for maximum correspondence over a comparison window, or the region designated as measured using one of the following sequence comparison algorithms or by manual alignment and visual inspection. Such sequences are then said to be "substantially identical." This definition also refers to the complement of a test sequence. Optionally, the identity exists over a region that is at least about 50 nucleotides in length, or more preferably over a region that is 100 to 500 or 1000 or more nucleotides in length. The term "similarity" or "likeness" percent, in the context of two or more polypeptide sequences, refers to two or more sequences or subsequences that have a specified percentage of amino acid residues that are either the same or similar. as defined in the 8 conservative amino acid substitutions defined above (ie, 60%, optionally 65%, 70%, 75%, 80%, 90% or 95% similar over a specified region or the complete sequence of a polynucleotide, for example, of the complete polypeptide sequences of the invention such as CCX-CKR2 (eg, SEQ ID NO: 2) or the extracellular domains of the polypeptides of the invention, when compared and aligned for maximum correspondence over a comparison window, or the region designated as it is measured using one of the following sequence comparison algorithms or through manual alignment and visual inspection.Such sequences are then said to be "s usually similar ". Optionally, this identity exists over a region that is at least about 50 amino acids in length, or more preferably over a region that is at least about 100 to 500 or 1000 or more amino acids in length. For sequence comparison, typically one sequence acts as a reference sequence, to which the test sequences are compared. When a sequence comparison algorithm is used, the reference test sequences are entered into a computer, the coordinates of the subsequences are designated, if necessary, and the parameters of the sequence algorithm program are designated. You can use error program parameters, or alternative parameters can be designated. The sequence comparison algorithm then calculates the percent of sequence identities for the test sequences relative to the reference sequence, based on the parameters of the program. A "comparison window", as used herein, includes reference to a segment of any of the number of contiguous positions selected from the group consisting, for example, of a full-length sequence of 20 to 600, approximately 50 to about 200 or about 100 to about 150 amino acids or nucleotides in which a sequence can be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned. Methods of sequence alignment for comparison are well known in the art. Optimal alignment of sequences for comparison can be conducted, for example, by the local homology algorithm of Smith and Waterman (1970) Adv. Appl. Math. 2: 482c, or by the homology alignment algorithm of Needleman and Wunsch (1970) J. Mol. Biol. 48: 443, by searching for the similarity method of Pearson and Lipman (1988) Proc. Nat '1 Acad, Sci USA 85: 2444, through computerized implementations of its algorithms (GAP, BESTFIT, FASTA and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr., Madison, Wl), or by manual alignment and visual inspection (see, for example, Ausubel et al., Current Protocols in Molecular Biology (1995 supplement)). An example of an algorithm that is suitable for determining percent sequence identity and sequence similarity are the BLAST and BLAST2.0 algorithms, which are described in Altschul et al., (1977) Nuc. Acids Res. 25: 3389-3402 and Altschul et al., (1990) J.
Mol. Biol. 215: 403-410 respectively. The software to analyze BLAST analyzes is publicly available through the National Center for Biotechnology Information (http://www.ncbi.nlm.nih.gov/). This algorithm involves first identifying high register sequence pairs (HSPs) by identifying short words of length W in the question sequence, which either matches or satisfies some positive value threshold T register when it is aligned with a word of the same length in a database sequence. T is referred to as the close word registration threshold (Altschul et al., Supra). These initial near word hits act as beginnings to initiate searches to find longer HSPs that contain them. Word hits extend in both directions along each sequence as soon as the cumulative alignment record can be incremented. The cumulative records are calculated using, for nucleotide sequences, the parameters M (record renumbered for a pair of equalization residuals, always <0) and N (record of penalty for record of unequalization, always <0). For amino acid sequences such as a register matrix is used to calculate the cumulative record. The extension of word hits in each direction is interrupted when: the alignment record, cumulative fails by the amount of X of its maximum achieved value; the cumulative record is 0 or lower, due to the accumulation of one or more alignments of negative record residues; or the end of any sequence is reached. The BLATS algorithms W, T and X determine the sensitivity and speed of the alignment. The BLASTN program (for nucleotide sequences) uses as errors a word length (W) of 11, such as an expectation (E) or 10, M = 5, N = -4 and a comparison of both strands. For amino acid sequences, the BLASTP program uses as errors a word length of 3, and the expectation (E) of 10, and the registration matrix BLOSUM62 (see, Henikoff and Henikoff (1989) Proc. Acad. Sci. USA 89 : 10915) alignments (B) of 50, expectation (E) of 10, M = 5, N = -4 and a comparison of both strands. The BLAST algorithm also performs a statistical analysis of the similarity between two sequences (see, for example, Karlin and Altschul (1993) Proc. Nati, Acad. Sci. USA 90: 5873-5787). A measure of similarity provided by the BLAST algorithm is the probability of smaller sums (P (N)), which provides an indication of the probability by which an equalization of two nucleotide or amino acid sequences would occur by chance. For example, a nucleic acid is considered similar to a reference sequence if the sum probability is smaller in a comparison of the test nucleic acid to the reference nucleic acid is less than about 0.2, more preferably less than 0.01, and much more preferably less than about 0.001. An indication that two acid sequences The nucleic acid or polypeptides are substantially identical in that the polypeptide encoded by the first nucleic acid is immunologically cross reactive with the antibodies raised against the polypeptide encoded by the second nucleic acid, as described below. So, a The polypeptide is typically substantially identical to a second polypeptide, for example, where the two peptides differ only by conservative substitutions. Another indication that two nucleic acid sequences are substantially identical is that the two molecules or their . supplements hybridize with each other and under severe conditions, as described below. Another indication that two nucleic acid sequences are substantially identical is that the primers can be used to amplify the sequence. "Modulators" of the activity of CCX-CKR2 are used to refer to molecules that increase or decrease the activity of CCX-CKR2 directly or indirectly and include those molecules identified using in vitro or in vivo assays for the link or signaling of CCX-CKR2. The activity of CCX-CKR2 can be increased, for example, by contacting the CCX-CKR2 polypeptide with an agonist, and / or in some cases, by expressing CCX-CKR2 in a cell. Agonists refer to molecules that increase the activity of CCX-CKR2. Agonists are agents that, for example, bind to, and stimulate, increase, open, activate, facilitate, increase activation, sensitize or up-regulate the activity of CCX-CKR2. Modulators can compete for binding to CCX-CKR2 with known CCX-CKR2 ligands such as SDF-1 and I-TAC and small molecules as described herein. Antagonists refer to molecules that inhibit CCX-CKR2 activity, for example, by blocking the binding of agonists such as I-TAC or SDF-1. Antagonists are agents that, for example, bind to, partially or totally block stimulation, decrease, prevent, delay activation, inactivate, desensitize, or regulate toward CCX-CKR2 activity. Modulators include agents that alter the interaction of CCX-CKR2 with: proteins that bind activators or inhibitors, receptors, including receptors coupled to G proteins (GPCRs), kinases, etc. Modulators include genetically modified versions of naturally occurring chemokine receptor ligands, for example, with altered activity, as well as naturally occurring and synthetic ligands, antagonists, agonists, small chemical molecules, siRNAs and the like. Assays for inhibitors and activators include, for example, the application of putative modulator compounds to a cell expressing CCX-CKR2 and then in determining functional effects on CCX-CKR2 signaling, eg, ERK1 and ERK2 phosphorylation or activation. members of the signal transduction pathways ERK1 or ERK2, and / or other effects as described herein. les or assays comprising CCX-CKR2 that are treated with a potential activator, inhibitor or modulator are compared to control les without the inhibitor, activator or modulator to examine the degree of inhibition. The control les (not treated with inhibitor) are assigned with a chemokine receptor activity value of 100%. Inhibition of CCX-CKR2 is achieved when the activity or expression value of CCX-CKR2 relative to the control is less than about 95%, optionally about 90%, optionally about 50% or about 25-0% . Activation of CCX-CKR2 is achieved when the activity or expression value of CCX-CKR2 relative to the control of at least about 105%, about 110%, optionally at least about 105%, about 150%, optionally so less about 105%, about 200-500%, or at least about 105%, about 1000-3000% or higher. "siRNA" refers to small interfering RNAs, which are capable of causing interference with expression and can cause post-transcriptional silencing of cell-specific genes, eg, mammalian cells (including human cells) and in the body, by example, mammalian bodies (including humans). The phenomenon of RNA interference is described and discussed in, Bass, Nature 411: 428-29 (2001); Elbahir et al., Nature 411: 494-98 (2001) and Fire et al., Nature 391: 806-11 (1998); and WO 01/75164, where the methods for making the interference RNA are also discussed. The siRNAs based on the sequences and nucleic acids encoding the gene products disclosed herein typically have less than 100 base pairs and can be, for example, about 30 BPS or shorter, and can be made by methods known in the art. the technique, including the use of complementary DNA strands or synthetic procedures. The siRNAs are capable of causing interference and can cause post-transcriptional silencing of specific genes in cells, e.g., mammalian cells (including human cells) and in the body, for example, mammalian bodies (including humans). Exemplary siRNAs according to the invention could have up to 29 bps, 25 bps, 22 bps, 21 bps, 20 bps, 15 bps, 10 bps, 5 bps or any integer around it or between them. The tools for designing optimal inhibitory siRNAs include those available from DNAengine Inc. (Seattle, WA) and from Ambion, Inc. (Austin, TX). An RNAi technique employs genetic constructs within which antisense sense sequences are placed in flanking regions to an intron sequence in an appropriate splice orientation with donor and acceptor splice sites. Alternatively, the spacing sequences of various lengths can be used to separate self-complementary regions of sequence in the construction. During the processing of the gene construction transcript, the intron sequences are spliced, allowing the sense and anti-sense sequences, as well as the splice junction sequences, to bind the formation of double-stranded RNA. The selection of ribonucleases then binds and segments the double-stranded RNA, in order to initiate the cascade of events that lead to the degradation of specific mRNA gene sequences and the silencing of specific genes. The term "compound" refers to a specific molecule and includes its enantiomers, diastereomers, polymorphs and salts thereof. The term "heteroatom" refers to a nitrogen, oxygen or sulfur bonded atom. The term "substituted" refers to a group that is linked to a molecule or group of origin. Thus, a benzene ring having a methyl substituent is a benzene substituted with methyl. Similarly, a benzene ring having 5 hydrogen substituents would be an unsubstituted phenyl group when linked to a parent molecule. The term "substituted heteroatom" refers to a group where a heteroatom is substituted. The heteroatom can be substituted with a group or atom, including, but limited to hydrogen, halogen, alkyl, alkylene, alkenyl, alkynyl, aryl, amylene, cycloalkyl, cycloalkylene, heteroaryl, heteroarylene, heterocyclyl, carbocycle, hydroxy, alkoxy, aryloxy and sulfonyl. Representative substituted heteroatoms include, by way of example, cyclopropyl aminyl, isopropyl aminyl, and benzyl aminyl and phenoxy. The term "alkyl" refers to a monovalent saturated hydrocarbon group which may be linear or branched. Unless defined otherwise, such alkyl groups typically contain from 1 to 10 carbon atoms. Representative alkyl groups include, by way of example, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, n-pentyl, n-hexyl, n-heptyl, n-octyl , n-nonyl, n-decyl and the like. The term "alkylene" refers to a divalent saturated hydrocarbon group which may be linear or branched. Unless defined otherwise, such alkylene groups typically contain from 1 to 10 carbon atoms. Representative alkylene groups include, by way of example, methylene, ethane-1,2-diyl ("ethylene"), propane-1,2-diyl, propane-1,3-diyl, butane-1,4-diyl, pentane-1,5-diyl and the like. The term "alkenyl" refers to a monovalent unsaturated hydrocarbon group which may be linear or branched having at least one, and typically 1, 2 or 3, carbon-carbon double bonds. Unless defined otherwise, such alkenyl groups typically contain from 2 to 10 carbon atoms. Representative alkenyl groups include, by way of example, ethenyl, n-propenyl, isopropenyl, n-but-2-enyl, n-hex-3-enyl and the like. The term "alkynyl" refers to a monovalent unsaturated hydrocarbon group which may be linear or branched and having at least one, and typically 1, 2 or 3, triple carbon-carbon bonds. Unless defined otherwise, such alkynyl groups typically contain from 2 to 10 carbon atoms. Representative alkynyl groups include, by way of example, ethynyl, n-propynyl, n-but-2-ynyl, n-hex-3-ynyl and the like. The term "aryl" refers to a monovalent aromatic hydrocarbon having a single ring (ie, phenyl) or fused rings (ie, naphthalene). Unless defined otherwise, such aryl groups typically contain 6 to 10 carbon atoms in the ring. Representative aryl groups include, by way of example, phenyl and naphthalen-2-yl and the like. The term "arylene" refers to a divalent aromatic hydrocarbon having a single ring (ie, phenylene) or fused ring (ie, naphthalendiyl). Unless defined otherwise, such an ilene groups typically contain from 6 to 10 carbon atoms in the ring. Representative amylene groups include, by way of example 1, 2-phenylene, 1,3-phenylene, 1,4-phenylene, 5-diyl, naphthalene-2,7-diyl and the like. The term "aralkyl" refers to an aryl group substituted with alkyl. Representative aralkyl groups include benzyl. The term "cycloalkyl" refers to a monovalent saturated carbocyclic hydrocarbon group having a single ring or fused rings. Unless defined otherwise, such cycloalkyl groups typically contain from 3 to 10 carbon atoms. Representative cycloalkyl groups include, by way of example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like. The term "cycloalkylene" refers to a divalent saturated carbocyclic hydrocarbon group having a single ring or fused rings. Unless defined otherwise, such cycloalkylene groups typically contain from 3 to 10 carbon atoms. Representative cycloalkylene groups include, by way of example, cyclopropane-1,2-diyl, cyclobutyl-1,2-diyl, cyclobutyl-1,3-diyl, cyclopentyl-1,2-diyl, cyclopentyl-1,3-diyl , cyclohexyl-1,2-diyl, cyclohexyl-1,3-diyl, cyclohexyl-1,4-diyl and the like. The term "heteroaryl" refers to a substituted or unsubstituted monovalent aromatic group having a single fused ring or rings and containing in the ring at least one heteroatom (typically from 1 to 3 heteroatoms) selected from hydrogen, oxygen or sulfur . Unless defined otherwise, such heteroaryl groups typically contain from 5 to 10 total ring atoms. Representative heteroaryl groups include, by way of example, monovalent pyrrole, imidazole, thiazole, oxazole, furan, thiophene, triazole, pyrazole, isoxazole, isothiazole, pyridine, pyrazine, pyridazine, pyrimidine, triazine, indole, benzofuran, benzothiophene, benzimidazole, benzthiazole, quinoline, isoquinoline, quinazoline, quinoxaline and the like, where the point of attachment is any atom in the available carbon or nitrogen ring. The term "heteroarylene" refers to a divalent aromatic group having a single ring or fused rings and containing at least one heteroatom (typically 1 to 3 heteroatoms) selected from nitrogen, oxygen or sulfur in the ring. Unless defined otherwise, such heteroarylene groups typically contain from 5 to 10 total ring atoms. Representative heteroarylene groups include, by way of example, bivalent pyrrole, imidazole, thiazole, oxazole, furan thiophene, triazole, pyrazole, isoxazole, isothiazole, pyridine, pyrazine, pyridazine, pyrimidine, triazine, indole, benzofuran, benzothiophene, benzimidazole species. , benzthiazole, quinoline, isoquinoline, quinazoline, quinoxaline and the like, where the point of attachment in any atom in the available carbon or nitrogen ring. The terms "heterocyclyl" or "heterocyclic group" refers to a saturated or unsaturated group (non-aromatic) monovalent, substituted or unsubstituted which has a single ring or multiple fused rings and which contains in the ring at least one heteroatom (typically from 1 to 3 heteroatoms) selected from nitrogen, oxygen or sulfur. Unless defined otherwise, such heterocyclic groups typically contain from 2 to 9 total ring atoms. Representative heterocyclic groups include, by way of example, monovalent species of pyrrolidine, morpholine, imidazolidine, pyrazolidine, piperidine, 1,4-dioxane, thiomorpholine, piperazine, 3-pyrroline and the like, where the point of attachment at any atom in the the carbon or nitrogen ring available. The term "carbocycle" refers to an aromatic or non-aromatic ring in which each atom in the ring is carbon. Representative carbocycles include cyclohexane, cyclohexene, and benzene. The terms "halo" or "halogen" refer to fluoro- (-F), chloro- (-C1), bromo- (-Br) and iodo- (-1). The term "hydroxy" or "hydroxyl" refers to an -OH group. The term "alkoxy" refers to a group -OR, where R can be an unsubstituted or substituted alkyl, alkylene, cycloalkyl or cycloalkylene. Suitable substituents include halo, cyano, alkyl, amino, hydroxy, alkoxy and amido. Representative alkoxy groups include, by way of example, methoxy, ethoxy, isopropyloxy and trifluoromethoxy. The term "aryloxy" refers to a group -OR, where R can be a substituted or unsubstituted aryl or heteroaryl group. Representative aryloxy groups include phenoxy.
The term "sulfonyl" refers to a group -S (0) 2R, where R can be alkyl, alkylene, alkenyl, alkynyl, aryl, cycloalkyl, cycloalkylene, heteroaryl, heteroarylene, heterocyclic or halogen. Representative sulfonyl groups include, by way of example, sulfonate, sulfamide, sulfonyl halides and dipropylamide sulfonate. The term "condensation" refers to a reaction in which two or more molecules are covalently linked. In the same way, the condensation products are the products formed by the condensation reaction. DETAILED DESCRIPTION OF THE INVENTION I. Introduction The present invention provides the discovery that the orphan receptor RDC1, referred to herein as CCX-CKR2, binds the chemokine ligands SDF1 and I-TAC. In addition, the present invention provides the surprising discovery of the involvement of CCX-CKR2 in cancer. Thus, the invention provides methods for diagnosing cancer by detecting CCX-CKR2. The invention also provides methods for inhibiting cancer by administering a modulator of. CCX-CKR2 to an individual with cancer. II. Polypeptides and Polynucleotides CCX-CKR2 In numerous embodiments of the present invention, nucleic acids encoding CCX-CKR2 polypeptides of interest will be isolated and cloned using recombinant methods. Such embodiments are used, for example, to isolate polynucleotides CCX-CKR2 (eg, SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NOY and SEQ ID NO: 9)) for the expression of protein or during the generation of variants, derivatives, expression cassettes or other sequences derived from a CCX-CKR2 polypeptide (eg, SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6., NO SEQ ID NO: 8 and SEQ ID N0: 10)), to monitor the expression of the CCX-CKR2 gene, for the isolation or detection of CCX-CKR2 sequences in different species, for diagnostic purposes in a patient, for example, for detecting mutations in CCX-CKR2 or to detect Xa expression of CCX-CKR2 nucleic acids or CCX-CKR2 polypeptides. In some embodiments, the sequences encoding CCX-CKR2 are operably linked to a heterologous promoter. In some embodiments, the nucleic acids of the invention are of any mammal, including, in particular, for example, a human, a mouse, a rat, a dog, etc. In some cases, the CCX-CKR2 polypeptides of the invention comprise the extracellular amino acids of the human CCX-CKR2 sequence (e.g., SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8 and SEQ ID NO: 10)) while other residues are either altered or absent. In other embodiments, the CCX-CKR2 polypeptides comprise ligand binding fragments CCX-CKR2. For example, in some cases, the fragments are linked I-TAC and / or SDFl. The structure of seven trans-membrane receptors (of which CCX-CKR2 is one) are well known to those skilled in the art and therefore the trans-membrane domains can be easily determined. For example, easily available hydrophobicity algorithms can be found on the Internet in the Protein G Coupled (GPCRDB) receptor database, for example, http: // www. gpcr org. / 7tm / seq / DR / RDCl_HUMAN. TABDR.html or http: //www.gpcr.org/7tm/seq/vis/swac/P25106.html. This invention depends on routine techniques in the field of recombinant genetics. The basic texts disclosing the general methods for use in this invention include Sambrook et al., Molecular Cloning, A Laboratory Manual (3- edition 2001); Kriegler, Gene Transfer and Expression: A Laboratory Manual (1990); and Current Protocols in Molecular Biology (Ausubel et al., eds., 1994)). Appropriate primers and probes for identifying genes encoding CCX-CKR2 from mammalian tissues can be derived from the sequences provided herein (eg, SEQ ID NO: 1) for a general review of PCR, see, Innis et al. PCR Protocols: A Guide to Methods and Applications, Academic Press, San Diego (1990). III. Development of Specific Therapeutics Molecules that bind to CCX-CKR2, including modulators of CCX-CKR2 function ie agonists or antagonists or CCX-CKR2 activity agents, are useful for treating a number of mammalian diseases, including cancer. Diseases or conditions of humans or other species that can be treated with antagonists of a chemokine receptor or other inhibitors of chemokine function, include, but are not limited to, for example, carcinomas, gliomas, mesotheliomas, melanomas, lymphomas, leukemias, adenocarcinomas, breast cancer, ovarian cancer, cervical cancer, glioblastoma, leukemia, lymphoma, prostate cancer and Burkitt's lymphoma, head and neck cancer, colon cancer, colorectal cancer, non-small cell lung cancer, cancer of small cell lung, cancer of the esophagus, stomach cancer, pancreatic cancer, hepatobiliary cancer, gallbladder cancer, small bowel cancer, rectal cancer, kidney cancer, bladder cancer, prostate cancer, penile cancer, cancer of the urethral, testicular cancer, cervical cancer, vaginal cancer, uterine cancer, ovarian cancer, thyroid cancer, parathyroid cancer, c adrenal cancer, pancreatic endocrine cancer, carcinoid cancer, bone cancer, skin cancer, retinoblastomas, Hodgkin's lymphoma, non-Hodgkin's lymphoma (see, CANCER: PRINCIPIES AND PRACTICE (DeVita, VT et al., eds 1997) for additional cancers); as well as brain and neuronal dysfunction, such as Alzheimer's disease and multiple sclerosis; kidney dysfunction; rheumatoid arthritis; rejection of the cardiac graft; atherosclerosis; asthma; glomerulonephritis; contact dermatitis; inflammatory bowel disease; colitis; psoriasis; reperfusion injury; as well as other disorders and diseases described herein. Alternatively, a CCX-CKR2 agonist can be used to treat the disease, for example, in renal, cerebral or neuronal dysfunction as well as in cases where the mobilization of the stem cells is therapeutic. A. Methods for Identifying Zuimioquine Receptor Modulators A number of different classification protocols can be used to identify agents that modulate the level of activity or function of CCX-CKR2 in cells, particularly in mammalian cells, and especially in human cells. In general terms, the classification methods involve the classification of a plurality of agents to identify an agent that interacts with CCX-CKR2 (or 'an extracellular domain thereof), for example, through the link to CCX-CKR2, the prevention of a ligand (e.g., I-TAC and / or SDF1) of the CCX-CKR2 link or the CCX-CKR2 activation. In some embodiments, the agent binds CCX-CKR2 with at least about 1.5, 2, 3, 4, 5, 10, 20, 50, 100, 500 or 1000 times the affinity of the agent for another protein. 1. Chemotherapy Receptor Linkage Assays In some embodiments, modulators of CCX-CKR2 are identified by classifying molecules that compete with a CCX-CKR2 ligand such as SDF1 or I-CT. Those skilled in the art will recognize that there are a number of ways to conduct competitive analysis. In some embodiments, samples with CCX-CKR2 are pre-incubated with a labeled CCX-CKR2 ligand and then contacted with a potential competing molecule. The alteration (eg, a decrease) in the amount of the ligand bound to CCX-CKR2 indicates that the molecule is a potential CCX-CKR2 modulator. Preliminary classifications can be conducted by classifying agents capable of binding to a CCX-CKR2, as at least some of the agents thus identified are probably modelers of the chemokine receptor. Binding assays usually involve contacting CCX-CKR2 with one or more test agents and allowing sufficient time for the protein and test agents to form a binding complex. Any of the link complexes formed can be detected using any of a number of established analytical techniques. Protein binding assays include, but are not limited to, immunohistochemical binding assays, reflux cytometry, radioligand binding, europium-labeled ligand binding, biotin-labeled ligand binding, and other assays that maintain the conformation of CCX- CKR2. The chemokine receptor used in such assays can be naturally expressed, cloned or synthesized. For example, by contacting CCX-CKR2 with a potential agonist and by measuring the activity of CCX-CKR2, it is possible to identify those molecules that stimulate CCX-CKR2 activity. 2. Cells and Reagents The classification methods of the invention can be performed as in vitro or cell-based assays. In vitro assays are performed, for example, using membrane fractions or whole cells comprising CCX-CKR2. Cell-based assays can be performed on any of the cells in which CCX-CKR2 is expressed. The cell-based assays involve whole cells or cell fractions containing CCX-CKR2 to classify the binding or modulation of the CCX-CKR2 activity agent by the agent. Exemplary cell types that can be used according to the methods of the invention include, for example, any of the mammalian cells including leukocytes such as neutrophils, monocytes, macrophages, eosinophils, basophils, mast cells and lymphocytes, such as T cells. and B cells, leukemias, Burkitt's lymphomas, tumor cells, endothelial cells, fibroblasts, cardiac cells, muscle cells, breast tumor cells, ovarian cancer carcinomas, cervical carcinomas, glioblastomas, liver cells, kidney cells and cells neuronal, as well as fungal cells, including yeast. The cells can be primary cells or tumor cells or other types of immortal cell lines. Of course, CCX-CKR2 can be expressed in cells that do not express an endogenous reaction of CCX-CKR2. In some cases, fragments of CCX-CKR2, as well as protein fusions, can be used for classification. When molecules competing for binding with CCX-CKR2 ligands are desired, the CCX-CKR2 fragments used are fragments capable of binding the ligands (for example, capable of binding I-TAC or SDF1). Alternatively, any CCX-CKR2 fragment can be used as a target to identify molecules that bind CCX-CKR2. The CCX-CKR2 fragments can include any fragment of, for example, at least 20, 30, 40, 50 amino acids to a protein containing all but one amino acid of CCX-CKR2. Typically, the ligand binding fragments will comprise transmembrane regions and / or most or all of the extracellular domains of CCX-CKR2. 3. Signaling activity In some modes, the signaling activated by the CCX-CKR2 activation is used to identify modulators of CCX-CKR2. The chemokine receptor signaling activity can be determined in many ways. For example, signaling can be determined by detecting the adhesion of the cell mediated by the chemokine receptor. Interactions between chemokines and chemokine receptors can lead to rapid adhesion through the modification of integrin affinity and avidity. See, for example, Laudanna, Immunological Reviews 186: 37-46 (2002). Signaling can also be measured by determining, qualitatively and quantitatively, secondary messengers, such as cyclic AMPs or inositol phosphates, as well as phosphorylation or dephosphorylation events can also be monitored. See, for example, Premack et al., Nature Medicine 2: 1174-1178 (1996) and Bokoch, Blood 86: 1649-1660 (1995). The examples provide results demonstrating that the activity of CCX-CKR2 is mediated by the MAPK pathway, specifically CCX-CKR2 promotes the phosphorylation of the MAPK proteins ERK1 and ERK2. An exemplary native sequence ERK1 is provided in Access GenBank Accession No. p27361, an exemplary native sequence ERK2 is provided in GenBank Accession No. p28482. Thus, some tests are designed to detect a signal in the. MAPK route. The term "signal" as used with respect to the MAPK route refers to a component that is part of the route and / or some activity or manifestation associated with the route. The component can be any molecule (e.g., a protein) involved (e.g., produced, used or modified) in the MAPK signal transduction pathway. The signal in some cases is the detection of phosphorylation of the MAPK protein such as ERK1 or ERK2. Other - signals that can be detected include the components that are formed and / or use current-down phosphorylation of ERK1 or ERK2, as well as activities associated with the formation and use of these components. Some assays can be conducted to detect components that are formed and / or used upstream of the phosphorylation of ERK1 or ERK2, as well as activities associated with the production or use of such upstream components. Coo is indicated in the above, in the case of ERK1 or ERK2, the upstream components include, for example, Ras, MKKK (for example, c-Raf1, B-Raf and A-Raf) and an MKK (for example, MKKl and MKK2). Downstream components include, for example, the protein p90RSK, c-JUN, c-FOS, CREB and STAT. Thus, each of these components can be detected in certain assays and / or the modification (eg, phosphorylation) of these components. In addition, the profiling of gene expression and protein secretion could also be detected. Some of the classification assays that are provided involve the detection of ERK1 and / or ERK2 phosphorylation. The phosphorylation of these proteins is ligand (eg, SDF1, ITAC) dependent. Thus, assays and certain classification methods are conducted in the presence of ITAC or SDF1 to promote phosphorylation of ERK1 and ERK2. When the assays are conducted with ITAC and SDF1, the assay can be simplified using cells that do not express CXCR3 or CXCR4 for the reasons described above. The determination of whether ERK1 or ERK2 has been phosphorylated can be performed using various methods. One option is to lyse the cells after they have been incubated with a CCX-CKR2 ligand and a test agent. The resulting lysate can then be analyzed by Western blotting by electrophoresing the lysate to separate proteins on a gel and then by probing the gene with antibodies that specifically bind to the phosphorylated form of ERK1 or ERK2. Such antibodies are available from Cell Signaling Technologies MA. The total amount of ERK1 or ERK2 optionally present can be determined using antibodies that specifically bind both the phosphorylated and non-phosphorylated forms of ERK1 and ERK2. Additional details that consider this procedure are set forth in the examples. Another option that is suitable for high performance classification is to use an ELISA method using antibodies that specifically bind the phosphorylated forms of ERK1 and ERK2. The equipment for performing such assays in high throughput formats are also available from Cell Signaling Technologies of Beverly, MA (see, for example, PathScan ™ Phospho-p44 / 42 MAPK (T202 / Y204) Sandwich ELISA Kit). Western blotting and ELISA techniques could be used to detect the presence of proteins that are components of the MAPK path downstream of the phosphorylation of ERK1 and ERK2 using antibodies that specifically recognize the components that are involved in the pathway. For example, phosphorylation of p90RSK can be estimated using commercially available phosphorus-specific antibodies analogous to Western detection of phosphorylated ERK2 or ERK2. In addition, other events downstream of the CCX-CKR2 activation can also be monitored to determine the signaling activity. Downstream events include those activities or manifestations that occur as a result of the stimulation of a chemokine receptor. Exemplary downstream events include, for example, the changed state of a cell (eg, from normal to cancer cell or from cancer cell to non-cancerous cell). Cell responses include the adhesion of cells (e.g., to endothelial cells). The standardized signaling labels involved in angiogenesis (eg, VEGF-mediated signaling) can also be monitored for effects caused by modulators of CCX-CKR2. The chorioallantoic membrane (CAM) assays for example, can be used to analyze the effects on angiogenesis or, for example, the Miles assay to study the effects on vascular permeability. In another example, cell survival can be measured as a surrogate for CCX-CKR2 activity. As described in more detail in the examples, the expression of CCX-CR2 results in prolonged cell survival of cells expressing CCX-CR2 cultured under low serum conditions as compared to cells not expressing CCX -CR2 grown under the same conditions. A) Yes, the antagonism of CCX-CR2 is expected to reduce cell survival, while activation (for example, via agonists) is expected to increase cell survival. As a result, cell survival and apoptosis can serve as a reading for CCX-CR2 activity. A wide variety of cell death and apoptosis assays can be incorporated into classification methods to identify CCX-CR2 modulators. In general, assays of this type typically involve subjecting a population of cells to conditions that induce cell death or apoptosis, usually both in the presence and absence of a test compound that is the potential modulator of cell death or apoptosis. An assay is then conducted with the cells, or an extract thereof, to estimate what effect the test agent has on cell death or apoptosis by comparing the degree of cell death or apoptosis in the presence and absence of the test agent . Instead of analyzing cell death or apoptosis, the opposite type of assay can be performed, specifically the analysis of cell survival, as well as related activities such as cell growth and cell proliferation. Regardless of the particular type of assay, some assays are conducted in the presence of a ligand that activates CCX-CR2 such as I-TAC or SDF-1. A variety of different parameters that are characteristic of cell death and apoptosis can be analyzed in the present classification methods. Examples of such parameters include, but are limited to, monitoring of activation of cellular pathways for toxicological responses by the gene or analysis of protein expression, DNA fragmentation, changes in the composition of cell membranes, membrane permeability, activation of death receptor components or downstream signaling pathways (eg, caspases), generic stress responses, B NF-kappa activation and mitogen responses. In view of the role that CCX-CKR2 plays in reducing apoptosis, another procedure is to analyze the opposite of apoptosis and cell death, specifically to conduct classifications in which cell survival or cell proliferation is detected. Cell survival can be detected, for example, by monitoring the length of time the cell remains viable, the length of time that a certain percentage of original cells remain alive, or an increase in the number of cells. These parameters can be monitored visually using established techniques. Another assay for estimating apoptosis involves labeling cells with Annexin V (conjugated to Alexa Fluor (r) 488 dye) and Propidium iodide (pi) (Molecular Probes, Eugene Oregon). Pl, a red fluorescent nucleic acid binding dye, is impermeable to both living and apoptotic cells. Pl only marks the necrotic cells by staining tightly the nucleic acids in the cell. Annexin V takes advantage of the fact that apoptotic cells translocate phosphatidylserine (PS) to the outer surface of the cell. Annexin V is a human anticoagulant with high affinity for (PS). In apoptotic cells, but not living cells, they express PS on their outer surface. Annexin V (marked with Alexa Fluor (r) 488) marks these cells with green fluorescence. The cells are then analyzed in a fluorescence activated cell sorter (FACS) to estimate the fluorescence in the red and green channels: apoptotic cells (Annexin positive, negative PI) fluoresce only in the green channel; living cells (Annexin negative, negative PI) exhibit low fluorescence in both the red and green channels; and the necrotic or dead cells (Annexin posivo, Pl positive) are strongly positive in both of the red and green channels. Other classification methods are based on the observation that the expression of certain regulatory proteins is induced by the presence or activation of CCX-CR2. The detection of such proteins can thus be used to indirectly determine the activity of CCX-CR2. As described in more detail in the examples below, a series of ELISA investigations were conducted to compare the relative concentration of several secreted proteins in the cell culture medium for cells transfected with CCX-CR2 and non-transfected cells. Through these studies it was determined that CCX-CR2 induces the production of a number of diverse regulatory proteins, including growth factors, chemokines, metalloproteinases and metalloproteinase inhibitors. Thus, some of the classification methods that are provided involve determining whether a test agent modulates the production of certain growth factors, chemokines, metalloproteinases, and metalloproteinase inhibitors by CCX-CR2. In some cases, the assays are conducted with cells (or extracts thereof) that have been cultured under limited serum conditions since this was found to increase the production of proteins induced by CCX-CR2 (see examples). The following proteins are examples of various classes of proteins that were detected, as well as specific proteins within each class: (1) growth factors (e.g., GM-CSF); (2) chemokines (e.g., RANTES, MCP-1); (3) metalloproteinase (e.g., M P3-1); and (4) metalloproteinase inhibitor (e.g., TIMP-19) It is expected that other proteins in these various classes can also be detected.These particular proteins can be detected using a standard immunological detection method that is known in the art. procedure that is suitable for use in a high performance format, for example, are ELISAs that are conducted on multi-well plates.An ELISA kit to detect TIMP-1 is available from DakoCytomation (Product Code No. EL513) Additional examples of antibody suppliers that specifically bind the proteins listed above provide in the examples below: Proteins such as metalloproteinases that are enzymes can also be detected by known enzymatic assays.In other embodiments, the potential modulators of CCX- CK2 are tested for their ability to modulate cell adhesion. umor to monolayers of endothelial cells have been studied as a model of metastatic invasion (see, for example, Blood and Zetter, 1032, 89-119 (1990). These monolayers of endothelial cells mimic the lymphatic vasculature and can be stimulated with various cytokines and growth factors (eg, TNFalpha and IL-lbeta). Cells expressing CCX-CKR2 can be evaluated for the ability to adhere to this monolayer in both static addition assays as well as assays where cells are under flow conditions to mimic the strength of the vasculature in vivo. Additionally, assays for evaluating adhesion can also be performed in vivo (see, for example, von Andrian, U. H. Microcirculation, 3 (3): 287-300 (1996)). 4. Validation Agents that are initially identified by any of the above classification methods can be further tested to validate the apparent activity. Preferably such studies are conducted with suitable animal models. The basic format of such methods involves the administration of a guide compound identified during an initial classification to an animal that serves as an animal model for humans and then determining whether the disease (e.g., cancer) is in fact modulated and / or the disease or condition is lessened. The animal models used in the validation studies are generally mammals of any kind. Specific examples of suitable animals include, but are not limited to, primates, mice, rats and zebrafish. B. Agents that interact with CCX-CKR2 Modulators of CCX-CKR2 (eg, antagonists or agonists) can include, for example, antibodies (including monoclonal, humanized or other types of binding proteins that are known in the art), small organic molecules, siRNAs, CCX-CKR2 polypeptides or variants thereof, chemokines (including but not limited to SDF-1 and / or I-TAC) chemokine mimetics, chemokine polypeptides, etc. The agents tested as modulators of CCX-CKR2 can be any small chemical compound, or a biological entity, such as a polypeptide, sugar, nucleic acid or lipid. Alternatively, the modulators may be genetically altered versions, or peptidomimetic versions, of a chemokine or other ligand. Typically, the test compounds will be small chemical molecules and peptides. Essentially any chemical compound can be used as a potential modulator or ligand in the assays of the invention, although more often compounds that can be dissolved in organic aqueous solutions (especially based on DMSO are used). The assays are designed to classify large chemical libraries by automating the assay steps by providing compounds of any convenient source to the assays, which are typically run in parallel (for example, microtitre formats on microtitre plates in robotic assays). It will be appreciated that there are many suppliers of chemical compounds, including Sigma (St. Louis, MO), Aldrich (St. Louis, MO), Sigma Aldrich (St. Louis, MO), Fluka Chemika-Biochemica Analytika (Buchs, Switzerland) and the similar ones. In some embodiments, the agents have a molecular weight of less than 1,500 daltons, and in some cases less than 1,000, 800, 600, 500 or 400 daltons. The relatively small size of the agents may be desirable because smaller molecules have a higher probability of having physiochemical properties compatible with good pharmacokinetic characteristics, including oral absorption of higher molecular weight agents. For example, the least likely agents to be successful as drugs based on permeability and solubility were described by Lipinski et al., As follows: they have more than 5 H-bond donors (expressed as the sum of OHs and NHs); which has a molecular weight over 500; have a LogP above 5 (or MLogP above 4.15); and / or having more than 10 H-bond receptors (expressed as the sum of Ns and Os). See, for example, Lipinski et al., Adv Drug Delivery Res 23: 3-25 (1997). Classes of compounds that are substrates for biological transporters are typically exceptions to the rule. In one embodiment, high throughput screening methods involve providing a combinatorial peptide or chemical library containing a large number of potential therapeutic compounds (modular potential or compounds of). Such "combinatorial chemical libraries" or "ligand libraries" then classify one or more assays, as described herein, to identify those members of the library (particularly chemical species or subclasses) that exhibit a desired characteristic activity. The compounds thus identified can serve as conventional "guide compounds" or by themselves can be used as potential or current therapeutics. A combinatorial chemical library is a collection of various chemical compounds generated either by chemical synthesis or biological synthesis, by combining a number of chemical "building blocks". For example, a linear combinatorial chemical library such as a polypeptide library is formed by combining a set of chemical building blocks (amino acids) in each possible way for a given compound length (i.e., the number of amino acids in a compound of polypeptide). Millions of chemical compounds can be synthesized through such combinatorial mixing or chemical building blocks. The preparation and classification of combinatorial chemical libraries is well known to those skilled in the art. Such combinatorial chemistry libraries include, but are not limited to, peptide libraries (see, for example, U.S. Patent 5,010,175, Furia, Int. J. Pept. Prot. Res. 37: 487-493 (1991) and Houghton et al., Nature 354: 84-88 (1991)). Other chemical substances to generate libraries of chemical diversity can also be used. Such chemical substances include, but are not limited to: peptoids (e.g., PCT Publication No. WO 91/19735) encoded peptides (e.g., PCT Publication No. WO 93/20242), random bio-oligomers (e.g. , PCT Publication No. WO 92/00091), 'benzodiazepines (eg, U.S. Patent No. 5,288,514), diversomers such as hydantoins, benzodiazepines and dipeptides (Hobbs et al., Proc. Nat. Acad. Sci. USA 90: 6909 -6913 (1993)), vinylogous polypeptides (Hagihara et al., J.
AMER. Chem. Soc. 114: 6568 (1992)), non-peptide peptidomimetics with glucose ladder (Hirschmann et al., J. Amer. Chem. Soc. 114: 9217-9218 (1992)), organic synthesis of small compound library analogues (Chen et al. collaborators, J. Amer. Chem. Soc. 116: 2661 (1994)), oligocarbamates (Cho et al., Sciense 261: 1303 (1993)), and / or peptidyl phosphonates (Campbell et al., J. Org. Chem. 59: 658 (1994)), nucleic acid libraries (see Ausubel, Berger and Sambrook, supra), libraries of peptide nucleic acid (see, for example, US Pat. No. 5,539,083), antibody libraries (see, for example, Vaughn et al., Nature Biotechnology, 14 (3): 309-314 (1996) and PCT / US96 / 10287), carbohydrate libraries, (see, for example, Liang et al., Science, 274: 1520-1522 (1996) and US Patent No. 5,593,853), organic molecule libraries small (see, for example, benzodiazepines, Baum C &EN, January 18, page 33 (1993), isoprenoids, U.S. Patent No. 5,569,588, thiazolidinones and metathiazanones, U.S. Patent No. 5,549,974, pyrrolidines, U.S. Patent Nos. 5,525,735, and 5,519,134; morpholino compounds, U.S. Patent No. 5,506,337; benzodiazepi nas, 5,288,514 and the like). Devices for the preparation of combinatorial libraries are commercially available (see, for example, 357 MPS, 390 MPS, Advance Chem Tech, Louisville KY, Symphony, Rainin, Woburn, MA, Applied Biosystems 433A, Foster City, CA, 9050 Plus, Millipore, Bedford, MA). In addition, numerous combinatorial libraries are commercially available by themselves (see, for example, ComGenex, Princeton, N.J., Tripos, St. Louis, MO, 3D Pharmaceutical, Exton, PA, Martek Biosciences, Colombia, MD, etc.). CCX7923 (see Figure 4) is commercially available and can be made by condensing N- [3- (dimethylamino) propyl] -N, N-dimethyl-1,3-propanediamine with bromomethyl-bicyclo (2, 2, 1). ) hept-2-ene by methods known in the art. CCX0803 (see Figure 4) is commercially available and can be made by the condensation of 3- (2-bromoethyl) -5-phenylmethoxy-indole and 2,4,6-triphenylpyridine by methods well known in the art. See, for example, Organic Function Group Preparations, 2nd Edition Volume 1, (S.R. Sandler &W. Karo 1983); Handbook of Heterocyclic Chemistry (A. R. Katritzky, 1985); Encyclopedia of Chemical Technology, 4- Edition (J. Kroschwitz, 1996). In one embodiment, the active compounds (ie, CCX-CKR2 modulators) of the present invention have the general structure (I): where m is an integer from 1 to 5 and each Y that replaces the benzyl ring is independently selected from the group consisting of hydrogen, alkyl, alkyl substituted with halo, alkylene, alkenyl, alkynyl, cycloalkyl, cycloalkylene, halogen, heterocyclic, aryl, arylene, heteroaryl, heteroarylene, hydroxy, alkoxy and aryloxy, n is 0, 1, 2 or 3; A is -CH- or -N-; R1 and R2 are each independently alkyl or hydrogen, or Z in combination with R1 and R2 form a 5- or 6- membered ring comprising at least one nitrogen and optionally comprising one or more additional heteroatoms, wherein the 5-6 membered ring is optionally and independently substituted with one or more selected portions of the group consisting of alkyl, alkenyl, phenyl, benzyl, sulfonyl and substituted heteroatom; R3, R4 and R5 are each independently selected from the group consisting of hydrogen, alkyl, substituted haloalkyl, alkylene, alkenyl, alkynyl, cycloalkyl, cycloalkylene, heterocyclic, aryl, arylene, heteroaryl, heteroarylene, hydroxy, alkoxy and aryloxy; and Rs is alkyl or hydrogen; with the proviso that if Z is nitrogen and R1 and R2 together with Z form a morpholinyl group, then n is 3, and at least one and at least one of R3, R4 and R5 is hydroxy, alkoxy or aryloxy; with the proviso that if n = 1, Z is carbon and R 1 and R 2 is combination is not -CH 2 CH 2 NCH 2 CH 2 -; or with the proviso that R1 together with R2 is -CH (CH3) (CH2) - then Z is -CH-; or with the proviso that if R5 is t-butyl then R3 is hydrogen; or with the proviso that if R4 and R5 together form a 5-membered ring, then at least one of the atoms attached to the phenyl ring is carbon. See, US Provisional Patent No. 60 / 434,912 filed December 20, 2002 and US Provisional Patent Application No. 60 / 516,151, filed December 20, 2003. The wavy bond connecting the olefin to the phenyl ring substituted means that the ring can be either cis or trans to R6. In a preferred embodiment, n is 1, 2 or 3. In another preferred embodiment, n is 2 or 3. In a further preferred embodiment, n is 3. In another embodiment, the preferred compounds have the general structure (I), wherein is R6 is hydrogen. In a further embodiment, the preferred compounds have the general structure (I), wherein R6 is methyl. In another embodiment, the preferred compounds have the general structure (I), wherein R3, R4 and R5 are independently hydrogen, hydroxy, alkyl, alkoxy, aryloxy and alkyl substituted with halo. More preferably, R3, R4 and R5 are independently alkoxy or hydrogen. In another embodiment, the preferred compounds have the general structure (I), wherein R4 is hydrogen and R3 and R5 are alkoxy (-OR), including trifluoroalkoxy groups such as trifluoromethoxy and (-OCH2CF3). In a further embodiment, R3 is hydrogen and R4 and R5 are alkoxy. Either of these embodiments, the alkoxy group can be methoxy (-OCH3) or ethoxy (-OCH2CH3).
In another embodiment, the preferred compounds have the general structure (I), where R4 and R5 together form a heterocyclic, aryl or heteroaryl ring. In another preferred embodiment, R3 is hydrogen and R4 and R5 together are -0 (CH2) 30-, - (CH) 4- or -N (CH) 2N-. In another embodiment, the preferred compounds have the general structure (I), wherein Z is nitrogen and Z in combination with R1 and R2 form a heteroaryl or heterocyclic group. In a preferred embodiment, the compounds have the general structure (I) m, Z is CH and Z in combination with R1 and R2 form a heteroaryl or heterocyclic group. The most preferable compounds have the general structure (I), where CH and Z in combination with R1 and R2 form a heterocyclic group containing nitrogen. In a further embodiment, Z in combination with R1 and R2 form a substituted or unsubstituted morpholinyl, pyrrolidinyl, piperidinyl or piperazinyl group. Preferred substituents for the heteroaryl or heterocyclic group include alkyl, alkenyl, alkynyl, cycloalkyl, aryl, aralkyl, heteroaryl, alkoxy, hydroxy, heteroatoms, and halides. In an especially preferred embodiment, the heteroaryl or heterocyclic groups is substituted with benzyl, phenyl, methyl, ethyl, cyclohexyl, methoxy-methyl (-CH2OCH3) or cyclohexyl-methyl (-CH2 (C6Hn)).
In one embodiment, a preferred compound has the general structure (I), wherein Z in combination with R1 and R2 is a group that pyrrolidinyl substituted with alkyl or methoxymethyl; a piperidinyl group substituted with benzyl, phenyl, methyl, ethyl or substituted heteroatoms; or a piperazinyl group substituted with benzyl, phenyl or sulfonyl. Especially preferred substituted heteroatom groups include alkoxy, aminyl, aminyl cycloalkyl, aminyl alkyl, aminyl cyclopropyl, aminyl isopropyl, aminyl benzyl and phenoxy. Preferably, the substituted heteroatom is in the 3-position of the piperidinyl ring. In another aspect, the preferred compounds have the general structure (I), wherein Z in combination with R1 and R2 is Preferred compounds having the general structure (I) can also have Z as a nitrogen atom, have R1 and R2 each as alkyl or methyl groups, or have R1 and R2 together forming -C (C (0) N (CH3) 2) (CH2) 3-. In another embodiment, Z in combination with R1 and R2 form a 5-membered ring including nitrogen and optionally including one or more additional heteroatoms. In this mode, n is preferably 1 and Z is preferably -CH-. In an especially preferred embodiment of this type, Z in combination with R1 and R2 is where R7 is preferably hydrogen, alkyl, aryl or aralkyl. In another preferred embodiment R7 can be a halogenated benzyl or phenyl group. In a further embodiment, R7 is preferably hydrogen, methyl, ethyl, benzyl or para-fluoro-phenyl. In another embodiment, the active compounds of the present invention have the general structure (II): where m is an integer from 1 to 5; each Y that replaces the benzyl ring is independently selected from the group consisting of hydrogen, alkyl, alkyl substituted with halo, alkylene, alkenyl, alkynyl, cycloalkyl, cycloalkylene, halogen, heterocyclic, arylene, heteroaryl, heteroarylene, hydroxy and alkoxy; n is 1, 2 or 3; and R3, R4 and R5 are each independently selected from the group consisting of hydrogen, alkyl, alkyl substituted by halo, alkylene, alkenyl, alkynyl, cycloalkyl, cycloalkylene, heterocyclic, aryl, arylene, heteroaryl, heteroarylene, hydroxy, alkoxy and aryloxy. As in structure (I) above, the wavy bond connecting the olefin to the substituted phenyl ring means that the ring can be either cis or trans.
In another embodiment, the preferred compounds may have the general structure (II), where n is 3. In another embodiment, the preferred compounds may have the general structure (II), wherein R3, R4 and R5 are substituted as described for Structure (I) above. Currently, the especially preferred compounds have the general structure (II), where R3, R4 and R5 are alkoxy or methoxy. While many synthetic routes known to those of ordinary skill in the art can be used to synthesize the active compounds of the present invention, a general synthesis method is given in Scheme I below.
Scheme I In Scheme I, the aldehyde (2) is subjected to a condensation reaction with the primary amine (3) via the reductive amination. Suitable primary amines are commercially available from Aldrich, Milwaukee, Wl, for example, or can be synthesized by chemical routes known to those of ordinary skill in the art. The tuning reaction can be carried out with reducing agent in any suitable solvent, including, but not limited to tetrahydrofuran (THF), dichloromethane or methanol to form intermediate (4). Suitable reducing agents for the condensation reaction include, but are not limited to, sodium cyanoborohydride (as described in Mattson et al., J. Org. Chem. 1990, 55, 2552 and Barney et al., Tetrahedron Lett. , 31, 5547); sodium triacetoxyborohydride (as described in 7? bdel-Magid et al., Tetrahedron Lett., 31: 5595 (1990)); sodium borohydride (as described in Gribble, Nutaitis Synthesis, 709 (1987)); iron pentacarbonyl and alcoholic KOH (as described in Watabane et al., Tetrahedron Lett., 1879 (1974)); and BH3-pyridine (as described in Welter et al., J. Chem. Soc. Perkin, Trnas. 1: 717 (1984)). The transformation of the intermediate (4) to the compound (5) can be carried out in any suitable solvent, such as tetrahydrofuran or dichloromethane, with an acyl chloride suitably substituted in the presence of a base. Tertiary amine bases are preferred. Especially preferred ones include triethylamine and Hunnings base. Alternatively, the transformation of the intermediate (4) to the compound (5) can also be obtained with a suitable coupling reagent, such as l-ethyl-3- (3-dimethylbutylpropyl) carbodiimide or Dicyclohexylcarbodiimide (as described in B. Neises and W. Steglich, Angew. Chem. Int. Ed. Engl. 17: 522 (1978)), in the presence of a catalyst, such as 4-N, N-dimethylamino-pyridine, or in the presence of hydroxybenzotriazole (as described in K. Synth Commun. 7: 251). To demonstrate that the compounds described in the above are useful antagonists for the chemokines SDF-1 and I-TAC, the compounds were classified in vi tro to determine their ability to displace SDF-1 and I-TAC of the CCX-CKR2 receptor in multiple concentrations. The compounds were combined with mammary gland cells expressing the CCX-CKR2 receptor sites in the presence of the chemokine SDF-1 labeled with 125 I and / or the chemokine I-TAC 125I. The ability of the compounds to displace the labeled SDF-1 and I-TAC from the CCX-CKR2 receptor sites at multiple concentrations was then determined with the classification process. Compounds that were considered to be effective SDF-1 and I-TAC antagonists were able to displace at least 50% of the chemokine SDF-1 and / or I-TAC of the CCX-CKR2 receptor in concentrations at or below 1.1 micromolar (μM) and more preferably at concentrations at or below 300 nanomolar (nM). In some cases, it is desirable that the compounds can displace at least 50% of the SDF-1 and / or I-TAC of the CCX-CKR2 receptor in concentrations at or below 200 nM. Exemplary compounds that meet these criteria are reproduced in Table I below. TABLE I C. High-Performance, Solid-Phase, Soluble Tests In the high-throughput assays of the invention, it is possible to classify up to several thousand different modulators or ligands in a single day. In particular, each cavity of a microtiter plate can be used to run a separate assay against a selected potential modulator, or, if the concentration or effects of the incubation time are to be observed, every 5-10 cavities can test a single modulator . Thus, a single standard microtiter plate can analyze approximately 100 (for example 96) modulators. If plates of 1536 cavities are used, then a single plate can easily analyze from about 100 to about 1500 different compounds. It is possible to analyze several different plates per day; Test classifications are possible for up to about 6,000-20,000 different compounds using the integrated systems of the invention. More recently, microfluidic procedures have been developed for the handling of reagents. The invention provides in vitro assays for identifying, in a high throughput format, compounds that can modulate the function or activity of CCX-CKR2. Control reactions that measure the CCX-CKR2 activity of the cell in a reaction that does not include a potential modulator are optional, since the assays are highly uniform. Such optional control reactions, however, increase the reliability of the assay. In some trials it will be desirable to have positive controls to ensure that the test components are working properly. Therefore, two types of positive controls are appropriate. First, a known activator or ligand of CCX-CKR2 can be incubated with a test sample, and the resulting increase in the signal resulting from increased activity of CCX-CKR2 (for example, as determined according to the methods in the present) . Second, an inhibitor or antagonist of CCX-CKR2 can be added, and the resulting decrease in the signal for chemokine receptor activity can be similarly detected. It will be appreciated that the modulators can also be combined with activators or inhibitors to find modulators that inhibit the increase or decrease that is otherwise caused by the presence of the known modulator of CCX-CKR2. IV. Expression of CCS-CKR2 in a subject In some embodiments, CCX-CKR2 is expressed in a subject, in order to thereby increase the expression of CCX-CKR2. Alternatively, the inhibitory polynucleotides, including, for example, siRNA or antisense sequences, may be expressed in vitro or in vivo to inhibit the expression of CCX-CKR2. In some cases, a polynucleotide encoding CCX-CKR2 is introduced into an in vitro cell and the cells are subsequently introduced into a subject. In some of these cases, the cells are first isolated from the subject and then reintroduced into the subject after the polynucleotide is introduced. In other embodiments, the polynucleotides encoding CCX-CKR2 are introduced directly into the cells in the subject in vivo. In some cases, the polypeptides encoding CCX-CKR2 are introduced into the cells of: (i) a tissue of interest, (ii) exogenous cells introduced into the tissue, or (iii) nearby cells that are not within the tissue. In some embodiments, the polynucleotides of the invention are introduced into endothelial cells. The tissue with which the endothelial cells are associated is any tissue in which it is desired to increase the migration or expansion of the endothelium. The conventional viral and non-viral base gene transfer methods can be used to introduce nucleic acids encoding designed polypeptides of the invention into mammalian cells or target tissues. Such methods can be used to deliver nucleic acids encoding polypeptides of the invention (eg, CCX-CKR2) to cells in vitro. In some embodiments, the nucleic acids encoding polypeptides of the invention are administered for gene therapy uses in vivo or ex vivo. Non-viral vector delivery systems include DNA plasmids, naked nucleic acid and nucleic acid formed in complex with a delivery vehicle such as a liposome. Viral vector delivery systems include DNA and RNA viruses, which have either episomal or integrated genomes after cell delivery. For a review of gene therapy procedure, see Anderson, KscienceK 256: 808-813 (1992); Nabel & Felgner, TIBTECH 11: 211-217 (1993; Mitani &Caskey, TIBTECH 11: 162-166 (1993); Dillon, TIBTECH 11: 167-175 (1993); Miller, Nature 357: 455-460 (1992); Van Brunt, Biotechnolpgy 6 (10): 1149-1154 (1988); Vigne, Restorative Neurology and Neuroscience 8: 35-36 (1995); Kremer & Perricaudet, Briish Medical Bulletin 51 (1): 31-44 (1995); Hadada and collaborators, in Current Topics in Microbiology and Immunology Doerfler and Bóhm (eds) (1995); and Yu et al, Gene Therapy 1: 13-26 (1994). Methods of non-viral delivery of nucleic acids encoding designed polypeptides of the invention include lipofection, microinjection, biolistics, virosomes, liposomes, immunoliposomes, polycation or lipid conjugates: nucleic acid, naked DNA, artificial virions and enhanced uptake by the DNA Lipofection is described in, for example, U.S. Patent Nos. 5,049,386, US 4,946,787; and US 4,897,355) and lipofection reagents are sold commercially (e.g., Transfectam ™ and Lipofectin ™). The cationic and neutral lipids are suitable for the efficient receptor recognition polyofeptide of polynucleotides include those of Fergner, WO 91/17424, WO 91/16024. The delivery can be to cells (ex vivo administration) or target tissues (administration in vivo). The preparation of lipid: nucleic acid complexes, including targeted liposomes such as immunolipid complexes, is well known to one skilled in the art (see, eg, Crystal, Science 270: 404-410). (nineteen ninety five); Baléese et al., Cancer Gene Ther. 2: 291-297 (nineteen ninety five); Behr et al., Bioconj ugate Chem. 5: 382-389 (1994); Remy et al., Bioconj ugate Chem. 5: 647-654 (1994); Gao et al., Gene Therapy 2: 710-722 (1995); Ahmad et al. Cancer Res. 52: 4817-4820 (1992); U.S. Patent Nos. 4,186,183, 4,217,344, 4,235,871, 4,261,975, 4,485,054, 4,501,728, 4,774,085, 4,837,028 and 4,946,787). The use of RNA-based or viral DNA systems for the delivery of nucleic acids encoding designed polypeptides of the invention takes advantage of highly evolved processes to direct a virus to specific cells in the body and transport the viral load to the nucleus. The viral vectors can be administered directly to patients (in vivo) or they can be used to treat cells in vitro and the modified cells are administered to patients (ex vivo). Conventional viral base systems for the polypeptide delivery of the invention could include retroviral, lentivirus, adenoviral, adeno-associated and herpes simplex virus vectors for gene transfer. Viral vectors are currently the most efficient and versatile method of gene transfer in target cells and tissues. Integration into the host genome is possible with retrovirus, lentivirus and adeno-associated virus gene transfer methods, often resulting in long-term expression of the inserted transgene. Additionally, high transduction efficiencies have been observed in many different cell types and target tissues. The tropism of a retrovirus can be altered by incorporating foreign envelope proteins, expanding the potential target population of the target cells. Lentiviral vectors are retroviral vectors that are capable of transducing or infecting non-dividing cells and typically produce high viral titers. The selection of a retroviral gene transfer system would therefore depend on the target tissue. Retroviral vectors are comprised of long terminal cis-acting repeats with packaging capacity for up to 6-10 kb of the foreign sequence. The minimal cis action LTRs are sufficient for the replication and packaging of the vectors, which are then used to integrate the therapeutic gene into the target cell to provide permanent transgene expression. Widely used retroviral vectors include those based on murine leukemia virus (MuLV), gibbon simian leukemia virus (GaLV), Simian Immunodeficiency Virus (SIV), human immunodeficiency virus (HIV) and combinations thereof (see, for example, Buchscher et al., J. Virol. 66: 2731-2739 (1992); Johann et al., J. Virol. 66: 1635-1640 (1992); Som erfelt et al., Virol. 176: 58-59 (1990); Wilson et al., J. Virol 63: 2374-2378 (1989); Miller et al., J. Virol. 65: 2220-2224 (1991); PCT / US94 / 05700). In applications where transient expression of the polypeptides of the invention is preferred, adenoviral based systems are typically used. Adenoviral-based vectors are capable of very high transduction efficiency in many cell types and do not require cell division. With such vectors, high titer and levels of expression have been obtained. This vector can be produced in large quantities in a relatively simple system. Adeno-associated virus vectors ("AAV") are also used to transduce cells with target nucleic acids, for example, in the in vivo production of nucleic acids and peptides and for gene therapy procedures in vivo and ex vivo (see, for example, example, West et al., Virology 160: 38-47 (1987), U.S. Patent No. 4,797,368, WO 93/24641, Kotin, Human Gene Therapy 5: 793-801 (1994), Myzyczka, J. Clin. : 1351 (1994)). The construction of recombinant AAV vectors are described in a number of publications, including U.S. Patent No. 5,173,414; Tratschin et al., Mol. Cell. Biol. 5: 3251-3260 (1985); Tratschi, et al., Mol. Cell. Biol. . 4: 2072-2081 (1984); Hermonat & Muzyczka, PNAS 81: 6466-6470 (1984); and Sa ulski et al., J. Virol. 63: 03822-3828 (1989). pLASN and MFG-S are examples of retroviral vectors that have been used in clinical experiments (Dunbar et al., Blood 85: 3048-305 (1995); Kohn and collaborators, Nat. Med. 1: 1017-102 (1995); and collaborators, PNAS 94:22 12133-12138 (1997)). PA317 / pLAS? was the first therapeutic vector used in a gene therapy experiment. (Blaese et al., Science 270: 475-480 (1995)). Transduction efficiencies of 50% or greater have been observed for vectors packed with MFG-S (Ellem et al., Immunol Immunother 44 (1): 10-20 (1997); Dranoff et al., Hum. Gene Ther.1: 111-2 (1997). Recombinant adeno-associated virus (rAAV) vectors are an alternative Promising gene delivery systems based on defective and non-pathogenic parvovirus adeno-associated virus type 2. All vectors are derived from a plasmid that retains only the AAV of 145 bp inverted terminal repeats flanking the transgene expression cassette. efficient gene transfer and stable transgene delivery due to integration into the transduced cell genomes are key factors for this vector system (Wagner et al., Lancet 351: 9117 1702-3 (1998), Kearns et al., Gene Ther 9: 748-55 (1996)) Replicating deficient recombinant adenoviral vectors (Ad) can be designed such that one transgene replaces the Ad Ela, Elb and E3 genes, subsequently the replication defector vector it is propagated in the human 293 cells that supply the function of the suppressed gene in trans. Ad vectors can transduce multiple tissue types in vivo, including differentiated, non-dividing cells such as those found in the tissues of the liver, kidney and muscular system. Conventional Ad vectors have a large carrier capacity. An example of using an Ad vector in a clinical experiment involved polynucleotide therapy for antitumor immunization with intramuscular injection (Sterman et al., Hum Gene 7: 1083-9 (1998)). Additional examples of the use of adenovirus vectors for gene transfer in clinical experiments include Rosenecker et al., Infection 24: 1 5-10 (1996); Sterman et al., Hum. Gene Ther. 9: 7 1083-1089 (1998); Welsh et al., Hum. Gene Ther. 2: 205-18 (nineteen ninety five); Alvarez et al., Hum. Gene Ther. 5: 597-613 (1997); Topf et al., Gene Ther. 5: 507-513 (1998); Sterman et al., Hum. Gene Ther. 7: 1083-1089 (1998). The packaging cells are used to form virus particles that are capable of infecting a host cell. Such cells include 293 cells, which package adenovirus, and? 2 cells or PA317 cells that package retroviruses. Viral vectors used in gene therapy are usually generated by the producer cell line that packages a nucleic acid vector into a viral particle. The vectors typically contain the minimal viral sequences required for packaging and subsequent integration into a host, other viral sequences that are replaced by an expression cassette for the protein that is expressed. Absent viral functions are supplied in trans through the packaging cell line. For example, AAV vectors used in gene therapy typically only possess ITR sequences from the AAV genome that are required for packaging and integration into the host genome. Viral DNA is packaged in a cell line, which contains a helper plasmid that encodes the other AAV genes, specifically rep and cap, but lacks ITR sequences. The cell line is also infected with adenovirus as an assistant. The helper virus promotes the replication of the AAV vector and the expression of AAV genes from the helper plasmid. The helper plasmid is not packaged in significant amounts due to a lack of ITR sequences. Adenovirus contamination can be reduced by, for example, heat treatment to which the adenovirus is more sensitive than AAV. In many gene therapy applications, it is desirable that the gene therapy vector be delivered with a high degree of specificity to a particular tissue type. A viral vector is typically modified to have specificity for a given cell type by expressing a ligand as a fusion protein with a viral coat protein on the outer surface of the virus. The ligand is selected to have affinity for a known receptor that is present in the type of cell of interest. For example, Han et al., PNAS 92: 9747-9751 (1995), reported that Moloney murine leukemia virus can be modified to express gp70 fused human heregulin, and the recombinant virus infects certain human breast cancer cells that express the receptor of human epidermal growth factor. This principle can be extended to other pairs of viruses that express a ligand fusion protein and the target cell that expresses a receptor. For example, filamentous phage can be designed to display antibody fragments (e.g., FAB or Fv) that have specific binding affinity for virtually any selected cellular receptor. Although the above description applies mainly to viral vectors, the same principles can be applied to non-viral vectors. Such vectors can be designed to contain specific uptake sequences although to favor uptake by specific target cells. Gene therapy vectors can be delivered in vivo by administration to an individual patient, typically by systemic administration (eg, intravenous, intraperitoneal, intramuscular, subdermal or intracranial infusion) or topical application, as described below. Alternatively, the vectors can be delivered to ex vivo cells, such as explanted cells from an individual patient (e.g., lymphocytes, bone marrow aspirates, tissue biopsy) or universal donor hematopoietic stem cells, followed by reimplantation of the cells into a patient, usually after selection for cells that have incorporated the vector. Transfection of cells ex vivo for diagnosis, research or for gene therapy (for example, by re-infusing the transfected cells in the host organism) is well known to those skilled in the art. In a preferred embodiment, the cells are isolated from the target organism, transfected with nucleic acid (gene or cDNA) encoding a polypeptide of the invention, and reinfused back into the target organism (eg, patient). Various types of cells suitable for ex vivo transfection are well known to those skilled in the art (see, for example, Freshney et al., Culture of Animal Cells, A Manual of Basic Technique (3rd ed., 1994)) and cited references in them for a discussion of how to isolate and grow cells from patients). In one embodiment, stem cells are used in ex vivo procedures for cell transfection and gene therapy. The advantage of using stem cells is that they can be differentiated into other cell types in vitro or they can be introduced into a mammal (such as the donor of the cells) where they will be grafted into the bone marrow. Methods for differentiating CD34 + cells in vitro in clinically important types of immune cells using cytokines such as a GM-CSF, IFN-γ. and TNF-a are known (see Inaba et al., J. Exp. Med. 176: 1693-1702 (1992)). The stem cells are isolated for transduction and differentiation using known methods. For example, stem cells are isolated from bone marrow cells by spreading bone marrow cells with antibodies that bind unwanted cells, such as CD4 + and CD8 + (T cells), CD45 + (panB cells), GR-1 (granulocytes) ) and Iad (cells presenting differentiated antigen) (see Inaba et al., J. Exp. Med. 176: 1693-1702 (1992)). Vectors (eg, retroviruses, adenoviruses, liposomes, etc.) containing therapeutic nucleic acid can also be administered directly into the body for cell transduction in vivo. Alternatively, the naked DNA can be administered. Administration and by any of the route normally used to introduce a final contact molecule or blood or tissue cells. Suitable methods of administration of such nucleic acids are available and well known to those skilled in the art., and although more than one route can be used to administer a particular composition, as a particular route it can often provide a more immediate and more effective reaction than another route.
The pharmaceutically acceptable carriers are determined in part by the particular composition that is administered, as well as by the particular method used to administer the composition. Accordingly, there is a wide variety of suitable formulations of pharmaceutical compositions of the present invention, as described below (see, for example, Remington's Pharmaceutical Sciences, 17 th ed., 1989). V. Diagnosis and Prognosis The present invention provides methods for detecting a cancer cell, including methods for providing a prognosis or diagnosis of cancer. As demonstrated herein, CCX-CKR2 is expressed in almost every cancer cell tested to date, whereas the normal (non-cancer) expression of CCX-CKR2 is presented to be limited to the kidney and some brain cells as well as in certain stages of fetal liver development. Therefore, expression of CCX-CKR2 in a cell, and in particular, in a non-fetal cell and / or a cell other than a kidney or brain cell, indicates the probable presence of a cancer cell. In some cases, samples containing cells expressing CCX-CKR2 are confirmed for the presence of cancer cells using other methods known in the art. According to yet another aspect of the invention, methods are provided for selecting a course of treatment of a subject having or suspected of having cancer. The methods include obtaining a biological sample from the subject, contacting the sample with antibodies or antigen-binding fragments thereof that binds specifically to CCX-CKR2, detecting the presence or absence of the antibody binding and selecting an appropriate course of treatment for the cancer of the subject. In some embodiments, the treatment is by administering CCX-CKR2 antagonists to the subject. Detection methods using agents that bind a protein are well known and include, for example, various immunoassays, flow cytometry, etc. Using flow cytometry, cells expressing a specific antigen of interest within a mixed population of cells can be identified. Briefly, the cells are allowed to react with an antibody specific for the protein of interest (eg, CCX-CKR2). The antibody can be either fluorescently labeled (direct spotting method), or if it is not labeled, a second antibody that reacts with the first one can be fluorescently labeled (indirect staining method). The cells are then passed through an instrument that can detect the fluorescent signal. The cells are aspirated and made into an individual cell suspension. This cell suspension is passed through a laser that excites the antibody labeled with fluorochrome that now binds the cells and acquires this data. Cells that are found to be bright (ie, react with the fluorescently labeled antibody) express the protein of interest; cells that are opaque (ie, do not react with the fluorescently labeled antibody) do not express the protein of interest. The present invention provides methods for diagnosing human diseases including, but not limited to cancer, for example carcinomas, gliomas, mesotheliomas, melanomas, lymphomas, leukemias, adenocarcinomas, breast cancer, ovarian cancer, cervical cancer, glioblastoma, leukemia, lymphoma, cancer of prostate, and Butkitt lymphoma, head and neck cancer, colon cancer, colorectal cancer, non-small cell lung cancer, small cell lung cancer, esophageal cancer, stomach cancer, pancreatic cancer, hepatobiliary cancer, gallbladder cancer, small bowel cancer, rectal cancer, kidney cancer, bladder cancer, prostate cancer, penile cancer, cancer of the urethra, testicular cancer, cervical cancer, vaginal cancer, uterine cancer, ovarian cancer, cancer thyroid cancer, parathyroid cancer, adrenal cancer, endocrine pancreatic cancer, carcinoid cancer, bone cancer, cancer of the the, retinoblastomas, Hodgkin's lymphoma, non-Hodgkin's lymphoma (see, CANCER: PRINCIPLES AND PRACTICE (DeVita, V.T. and collaborators. Eds. 1997) for additional cancers); as well as brain and neuronal dysfunction, such as Alzheimer's disease and multiple sclerosis; kidney dysfunction; rheumatoid arthritis; rejection of the cardiac graft, atherosclerosis; asthma; glomerulonephritis; contact dermatitis; inflammatory bowel disease; colitis; psoriasis; reperfusion injury; as other disorders and diseases described herein. In some modalities, the subject does not have Kaposi's sarcoma, Castleman's disease multicentric or primary effusion line associated with AIDS. As provided herein, including in the examples, normal and diseased cells and tissues can be distinguished on the basis of reactivity to an anti-CCX-CKR2 or SDF-1 and I-TAC monoclonal antibody. For example, cancer cells are detected by detecting in a cell a chemokine receptor for which SDF-la and I-TAC compete for binding. In addition, the difference in ligand binding between the chemokine results can be detected and such differences can be used to detect cells expressing CCX-CKR2. For example, no other chemokine receptor has both SDFl and I-TAC as ligands. The chemokine binding can be determined using tissue samples (e.g., biopsies) or can be directly monitored in a tissue in situ (e.g., using radiolabelled chemokine imaging). The immunoassays can also be used to qualitatively or quantitatively analyze CCX-CKR2.
A general review of the applicable technology can be found at Harlow & Lane, Antibodies: A Laboratory Manual (1988). Alternatively, non-antibody molecules with affinity for CCX-CKR2 can also be used to detect the receptor. Methods for producing polyclonal or monoclonal antibodies that specifically react with a protein of interest are known to those skilled in the art (see, for example, Coligan, Current Protocols in Immunology (1991); Harlow &Lane, Antibodies, A Laboratory Manual (1988), Goding, Monoclonal Antibodies: Principies and Practice (2d ed.1986), and Kohler and Milstein Nature, 256: 495-497 (1975)). Such techniques include the preparation of antibodies by selection of antibodies from libraries of recombinant antibodies in phage vectors or the like. For example, in order to produce antiserum for use in an immunoassay, the protein of interest or an antigenic fragment thereof, is isolated as described herein. For example, a recombinant protein is produced in a transformed cell line. An inbred race of mice, rats, guinea pigs or rabbits is immunized with the protein using a standard adjuvant, such as Freund's adjuvant and a standard immunization protocol. Alternatively, a synthetic peptide derived from the sequences disclosed herein and conjugated to a carrier protein can be used as an immunogen. An additional option is to use a cell expressing protein or a membrane or liposome fraction comprising CCX-CKR2 or a fragment thereof as an antigen. Antibodies raised against the cell, membrane fraction or liposome can then be selected for their ability to bind to the protein. The polyclonal sera are collected and titrated against the immunogen in an immunoassay, for example, a solid phase immunoassay with the immunogen immobilized on a solid support. Polyclonal antisera with a titer of 104 or greater are selected and tested for cross-reactivity against different and sometimes homologous proteins, using a competitive binding immunoassay. The specific monoclonal and polyclonal antibodies and antisera will usually bind to a KD of at least about 0.1 mM, more usually at least about 1 μM, preferably at least about 0.1 μM or better and much more preferably, 0.01 μM or better to CCX-CKR2. For the preparation of antibodies, for example, recombinant, monoclonal or polyclonal antibodies, many techniques known in the art can be used (see, for example, Kohler &Milstein, Nature 256: 495-497 (1975); Kozbor et al. Immunology Today 4:72 (1983); Colé et al., Pp. 77-96 in Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc. (1985); Coligan, Current Protocols in Immunology (1991); Harlow &La e, Antibodies, A Laboratory Manual (1988); and Goding, Monoclonal Antibodies: Principies and Practice (2d ed.1986)). The genes encoding the heavy and light chains of an antibody of interest can be cloned from a cell, for example, the genes encoding a monoclonal antibody can be cloned from a hybridoma and used to produce a recombinant monoclonal antibody. The libraries of genes encoding the heavy and light chains of monoclonal antibodies can also be made from the hybridoma or plasma cells. Random combinations of the heavy and light chain gene products generate a large accumulation of antibodies with different antigenic specificity (see, for example, Kuby, Immunology (3rd ed., 1997)). Techniques for the production of single chain antibody of recombinant antibodies (U.S. Patent 4,946,778, U.S. Patent No. 4,816,567) can be adapted to produce antibodies to the polypeptides of this invention. Also, transgenic mice, or other organisms such as other mammals, can be used to express humanized or human antibodies (see, for example, U.S. Pat. Nos. ,545,807; 5,545,806; 5,569,825; 5,625,126; 5,633,425; ,661,016; Marks et al., Bio / Technology 10: 779-783 (1992); Lonberg et al., Nature 368: 856-859 (1994); Morrison, Nature 368: 812-13 (1994); Fishwild et al., Nature Biotechnology 14: 845-51 (1996); Neuberger, Nature Biotechnology 14: 826 (1996); and Lonberg & Huszar, Jntern. .Rev. Immunol. 13: 65-93 (1995)). Alternatively, the phage display technology can be used to identify antibodies and heteromeric Fab fragments that specifically bind selected antigens (see, for example, McCafferty et al., Nature 348; 552-554 (1990); Marks et al., Biotechnology 10: 779-783 (1992)). Antibodies can also be made bispecific, that is, capable of recognizing two different antigens (see, for example, WO 93/08829, Traunecker et al., EMBO J. 10: 3655-3659 (1991); and Suresh et al. in Enzymology 121: 210 (1986)). Antibodies can also be heteroconjugates, for example, two covalently linked antibodies, or immunotoxins (see, for example, U.S. Patent No. 4,676,980, WO 91/00360, WO 92/200373, and EP 03089). Methods for humanizing or primatizing non-human antibodies are well known in the art. Such antibodies are useful for both detection and therapeutic applications. Generally, a humanized antibody has one or more amino acid residues introduced therein from a non-human source. These non-human amino acid residues are often referred to as import residues, which are typically taken from a variable import domain. Humanization can be essentially performed following the method of Winter et al. (See, for example, Jones et al., Nature 321: 522-525 (1986); Riechmann et al., Nature 332: 323-327 (1988); Verhoeyen et al. , Science 239: 1534-1536 (1988) and Presta, Curr. Op. Struct. Biol. 2: 593-596 (1992)), by substituting rodent CDRs or CDR sequences for the corresponding sequences of a human antibody. Accordingly, such humanized antibodies are chimeric antibodies (U.S. Patent No. 4,816,567), wherein substantially less than an intact human variable domain is replaced by the corresponding sequences from a non-human species. In practice, humanized antibodies are typically human antibodies in which some CDR residues and possibly some FR residues are replaced by residues from analogous sites in rodent antibodies. SAW. Methods of Treatment, Administration and Pharmaceutical Compositions. Modulators of CCX-CKR2 (eg, antagonists or agonists) can be administered directly to the mammalian subject for modulation of chemokine receptor signaling in vivo. In some modalities, the modulators compete with SDFl and / or I-TAC for the link to CCX-CKR2. Modulation of CCX-CKR2 can include, for example, antibodies (including monoclonal, humanized or other types of binding proteins that are known in the art), small organic molecules, siRNAs, etc. In some embodiments, modulators of CCX-CKR2 are administered to a subject having cancer. In some cases, modulators of CCX-CKR2 are administered to treat cancer, for example, carcinomas, gliomas, mesotheliomas, melanomas, lymphomas, leukemias, adenocarcinomas, breast cancer, ovarian cancer, cervical cancer, glioblastoma, leukemia, lymphoma, cancer of prostate, Burkitt's lymphoma, head and neck cancer, colon cancer, colorectal cancer, non-small cell lung cancer, small cell lung cancer, esophageal cancer, stomach cancer, pancreatic cancer, hepatobiliary cancer, cancer of the gallbladder, small bowel cancer, rectal cancer, kidney cancer, bladder cancer, prostate cancer, penile cancer, urethral cancer, testicular cancer, cervical cancer, vaginal cancer, uterine cancer, ovarian cancer, cancer thyroid, parathyroid cancer, adrenal cancer, endocrine pancreatic cancer, carcinoid cancer, bone cancer, skin cancer retinoblastomas, Hodgkin's lymphoma, li non-Hodgkin's disease (see, C NCER: PRINCIPLES AND PRACTICE (DeVita, V.T. and collaborators, eds. 1997) for additional cancers); as well as brain and neuronal dysfunction, such as Alzheimer's disease and multiple sclerosis; kidney dysfunction; rheumatoid arthritis; rejection of the cardiac graft; atherosclerosis; asthma; glomerulonephritis; contact dermatitis; inflammatory bowel disease; colitis, psoriasis; reperfusion injury; as well as other disorders and diseases described herein. In some embodiments, the subject does not have Kaposi's sarcoma, multicentric Castleman's disease, or primary effusion lymphoma associated with AIDS. Since CCX-CKR2 is frequently expressed in cancer cells but not in non-cancer cells, it is typically desirable to administer CCX-CKR2 antagonist to treat subjects who have cancer. In some cases, the modulators have a molecular weight of less than 1,500 daltons, and in some cases less than 1,000,800,600,500, or 400 daltons.
The administration of the modulators can be by any of the routes normally used to introduce a modulator compound in final contact with the tissue being treated and is well known to those skilled in the art. Although more than one route can be used to administer a particular composition, a particular route can often provide a more immediate and more effective reaction than another route. The pharmaceutical compositions of the invention may comprise a pharmaceutically acceptable carrier. The pharmaceutically acceptable carriers are determined in part by the particular composition that is administered, as well as the particular method used to administer the composition. Accordingly, there is a wide variety of suitable formulations of pharmaceutical compositions of the present invention (see, for example, Remington's Pharmaceutical Sciences, 17th ed., 1985)). Modulators (eg, agonists or antagonists) of the expression or activity of CCX-CKR2, alone or in combination with other suitable components, can be made in aol formulations (ie, they can be "nebulized") to be administered by the inhalation route. The aol formulations can be placed in acceptable pressurized propellants, such as dichlorodifluoromethane, propane, nitrogen and the like.
Formulations suitable for administration include aqueous and non-aqueous solutions, sterile isotonic solutions, which may contain antioxidants, regulatory solutions, bacteriostats and solutes that transform the isotonic solution, and sterile aqueous and non-aqueous suspensions which may include suspending agents, solubilizers, agents of thickening, stabilizers and preservatives. In the practice of this invention, the compositions may be administered, for example, orally, nasally, topically, intravenously, intraperitoneally or intrathecally. Compound formulations may be provided in sealed unit dose or multiple dose containers, such as ampoules and flasks. The solutions and suspensions can be prepared from sterile powders, granules and tablets of the kind previously described. Modulators can also be administered as part of a prepared food or drug. In some embodiments, the CCX-CKR2 modulators of the present invention can be administered in combination with other appropriate therapeutic agents, including, for example, chemotherapeutic agents, radiation, etc. The selection of appropriate agents for use in the combination therapy can be made by one of ordinary skill in the art, in accordance with conventional pharmaceutical principles. The combination of therapeutic agents can act synergistically to effect the treatment or prevention of various disorders such as, for example, cancer, kidney dysfunction, brain dysfunction or neuronal dysfunction. Using this procedure, therapeutic efficacy can be achieved with lower dosages of each agent, thereby reducing potential for adverse side effects. The dose administered to a patient, in the context of the present invention, should be sufficient to effect a beneficial response in the subject over time (eg, to reduce tumor size or tumor burden). The optimal dose level for any patient will depend on a variety of factors including the effectiveness of the specific modulator employed, age, body weight, physical activity and diet of the patient, in a possible combination with other drugs, and on the severity of a particular disease. The size of the dose will also be administered by the existence, nature and degree of any of the adverse side effects that accompany the administration of a particular compound or vector in a particular subject. In determining the effective amount of the modulator that is administered a physician can value the levels in the circulating plasma of the modulator, the toxicity to the modulator and the production of anti-modulator antibodies. In general, the equivalent dose of a modulator is from about 1 ng / kg to 10 mg / kg for a typical subject. For administration, the chemokine receptor modulators of the present invention can be administered at a rate determined by LD-50 of the modulator, and the side effects of the modulator at various concentrations, as applied to the mass and the overall health of the subject. . The administration can be carried out via individual or divided doses. VII. COMPOSITIONS, EQUIPMENT, INTEGRATED SYSTEMS AND PROTEOMIC APPLICATIONS The invention provides compositions, equipment and integrated systems for practicing the assays described herein using anti-CCX-CKR2 antibodies or other agents that specifically detect CCX-CKR2. The invention provides test compositions for use in assays or solid phase; such compositions may include, for example, a CCX-CKR2 polypeptide (including, for example, as part of a cell, membrane fractions or liposomes (see, for example, Babcok et al., J. Biol. Chem. 276 (42) : 38433-40 (2001); Mirzabekov et al., Nat. Biotechnol. 18 (6): 649-54 (2000)) immobilized on a solid support, and a labeling reagent. In each case, the test compositions may also include additional reagents that are desirable for hybridization. For example, the solid support can be, for example, a petri dish, multi-well plate or microarray. In addition, the microarrays of peptide libraries can be used to identify peptide sequences that specifically bind CCX-CKR2. Agents that specifically bind to CCX-CKR2 can be included in the test compositions. For example, an antibody that specifically binds CCX-CKR2 can be immobilized on a solid support. In some of these embodiments, the agent is used to detect in the presence or absence of CCX-CKR2 or cells expressing CCX-CKR2. For example, the solid support can be a petri dish, multiple cavity plate or microarray. The invention also provides equipment for carrying out the assays of the invention. The kits typically include an agent (e.g., an antibody or other small molecule) that specifically binds to CCX-CKR2 and a label to detect the presence of the agent. The kits may include one or more polypeptides of the chemokine receptor. The kits may include any of the compositions mentioned above, and optionally also include additional components such as instructions for practicing a high throughput method of analysis for an effect on the activity or function of the chemokine receptors, one or more containers or compartments (for example, to contain the probe, tags, or the like) a modulator to control the function or activity of the chemokine receptors, an armature Robotics to mix the components of the equipment or the like. In some modalities, the equipment includes SDFl and / or I-TAC. In some modalities, the equipment includes tagged or labeled SDF-1 and cold-competing I-TAC or alternatively-, a labeled or labeled I-TAC and cold competing SDF-1. The labeled or labeled chemokine can be labeled or labeled and in any manner known to those skilled in the art. In some embodiments, the labeled chemokine is radiolabelled or labeled with biotin or a fluorescent mark. Alternatively, or in addition, the kit may contain an anti-I-TAC binding reagent (e.g., an antibody) for the detection of I-TAC. The equipment may also contain the appropriate salt buffer solutions and other reagents to perform a test of . competitive binding, for example, on intact cells or cell membranes. Such reagents are described in, for example, the examples below. In some aspects, the kits also comprise a solid support or receptacle for measuring the ligand binding to CCX-CKR2 (e.g., in a plate format for reactions compatible with scintillation counters or automated plate readers). In some aspects, the equipment comprises instructions for the use of the equipment, for example, in the methods of the invention. The invention also provides integrated systems for the high throughput classification of potential modulators for an effect on the activity or function of the potential CCX-CKR2 modulators. Systems typically include a robotic armor that transfers fluids from one source to a destination, a controller that controls the robotic armor, a mark detector as a data storage unit that records the mark detection, and a test component such as a microtitre dish comprising a cavity having a reaction mixture to a substrate comprising a fixed nucleic acid or immobilization portion. The optical images displayed (and optionally recorded) by a camera or other recording device (eg, a photodiode and data storage device) are optionally further processed in any of the embodiments herein, for example, digitize the image and when storing and analyzing in the image on a computer. A variety of commercially available peripheral equipment and software is available to digitize, store and analyze a digitized video or digitized optical image. EXAMPLES Example 1: This example shows that SDF-1 and I-TAC compete for the link to a new chemokine receptor. Materials and Methods Reagents and Cells. Recombinant human, viral and murine chemokines were obtained from R & amp; amp; amp;; D Systems (Minneapolis, MN) and PeproTech (Rocky Hill, NJ) where indicated. SDF-labeled with 125 I was purchased from PerkinElmer Life Sciences, Inc. (Boston, MA) and I-TAC labeled with 125 I was obtained from Amersham Pharmacia Biotech (Buckinghamshire, UK). Monoclonal antibodies used in flow cytometry and ligand binding competition were from R &D Systems (Minneapolis, MN): anti-CXCR4 clones 12G5, 44708.111 (171), 44716111 (172), 44717111 (173), nmIgG2a and nmIgG2b.
The secondary antibody, goat anti-mouse IgG PE conjugate (Coulter Immunotech, Miami, FL), was used to detect antibody binding by flow cytometry. The following cells were obtained from the American Type Culture Collection (Manassas, VA): MCF-7 (adenocarcinoma, mammary gland); MDA MB-231 (adenocarcinoma, mammary gland), MDA MB-435s (ductal carcinoma, mammary gland), DU 4475 (mammary gland), ZR 75-1 (ductal carcinoma, mammary gland), HEK 293 (human embryonic kidney) , HUV-EC-C (human umbilical vein, vascular endothelium, normal). The CEM-NKr cells (acute lymphoblastic leukemia, peripheral blood, T lymphoblast) were obtained from the NIH AIDS Research and Reference Reagent Program. Cell lines were cultured in DMEM (Mediatech, Rendón, VA) supplemented with 10% fetal bovine serum (FBS) (HyClone Logan, UT) at 37 ° C in a humidified incubator in a 5% C02 / air mixture . Human peripheral blood mononuclear cells (PBMC). were obtained from lymph curds from healthy donors (Stanford Blood Center, Palo Alto, CA) by centrifugation in Ficoll-Hypaque density gradients. Isolated PBMC were activated with 2.5 ug / ml of phytohemagglutinin (PHA) (Sigma Chemical Company, St. Louis, MO) and 10 ng / ml of recombinant human IL-2 (R & D Systems, Minneapolis, MN) for 3 days in RPMI-1640 (Mediatech, Rendón, VA) supplemented with 10% FBS at 37 ° C in a humidified incubator in a 5% C02 / air mixture. After activation, the cells were washed and RPMI supplemented with 10% FBS and 10 ng / ml of IL-2 was grown and renewed every 3-4 days until the day the cells were used. Link analysis. The inventors used their technique, "DisplaceMax ™" to examine the overall profile of the interaction of chemokine ligand with the SDF1 receptor in MCF-7 and CEM-NKr cells. This technology employs maximized efficiency radioligand binding, expanded using filtration protocols as previously described (Dairaghi et al., J. Biol. Chem 273: 21569-74 (1999); Gosling, J. et al. J. Immunol 164: 2851-6 2000). In these assays, DisplaceMax ™ employed the simultaneous interrogation of MCF-7 or CEM-NKr cells, as indicated, by > 110 different purified chemokines in the ability to displace SDF-la or I-TAC, radiolabeled with 125I, as indicated, using the described protocol (Dairahi, et al., J Biol. Chem 274: 21569-74 (1999); Gosling, J. and coworkers J Immunol 164: 2851-6 (2000)).
Briefly, the chemokine elements were incubated with cells followed by the addition of radiolabelled chemokine (125 I SDF-la or 125 I h I-TAC) for 3 h at 4 ° C in the following binding medium (25 mM HEPES, 140 mM NaCl, 1 mM CaCl 2, 5 mM MgCl 2 and 0.2% bovine serum albumin, adjusted at pH 7.1). Small molecules included in some trials, where indicated. In these tests the compound was added to the plate at the indicated concentration followed by the addition of radiolabelled chemokine. All assays were then incubated for 3 h at 4 ° C with slight agitation. After incubation in all the binding assays, the reactions were aspirated on glass filters GF / B treated with PEI (Packard) using a cell harvester (Packard) and washed twice (25 mM HEPES, 50 mM NaCl, 1 mM CaCl 2, 5 mM MgCA, adjusted to pH 7.1). The scintillating agent (MicroScint 10, Packard) was added to the wells, and the filters were counted in a Packard Topcount scintillation counter. The data was analyzed and plotted using Prism (GraphPad Prism version 3.0a for Macintosh, GraphPad Software). Determination of the Receiver Link 125I SDF-la. Using the filtration-based assay described above, the cells were preincubated with either 1) regulatory solution alone, 2) excess SDF-1β (90 nM final) or 3) MIG (175 nM final) as indicated by 30 min at 4 ° C. After this incubation, the cold chemokine competitor indicated at established concentrations and 125 Ih I-TAC were added to binding reactions. All assays were then incubated, harvested and analyzed as described above. RT PCR. mRNA was isolated from cells using standard techniques. The complementary DNA was analyzed for the expression of CXCR3 and CXCR4 by PCR. The specific primers were obtained from Integrated DNA technologies (Coraville, IA). The specific PCR products were measured by means of a Hybaid Omn-E (E &K Scientific Products, Inc., Saratoga, CA) for 35 cycles. GAPDH was measured as a control. Adhesion test. HUVEC cells were grown overnight in platens treated with tissue culture in the presence of TNFa (25 ng / ml) and IFN? (50 ng / ml). The next day the NSO cells transfected with CCX-CKR2 as well as the wild-type controls were labeled with calcein-AM. The calcein-labeled cells were then plated onto the endothelial monolayer in the presence or absence of the CCX-CKR2 antagonist (CCS3451). The platens were incubated at 37 ° C for 40 minutes followed by washing with PBS to remove non-adherent cells. Adherent NSO cells were visualized by fluorescent microscopy. The cells treated with the compound or vehicle were counted with the naked eye from three fields of vision (fov) and plotted. Results Recent reports have identified the expression of CXCR4 in several types of tumor cells (Sehgal, et al., J Surg Oncol 69: 99-104 (1998); Sehgal, A., et al., J. Surg Oncol 69: 239- 48 (1998); Burger et al Blood 94: 3658-67 (1999), Rempel et al., Clin Cancer Res 6: 102-11 (2000); Koshiba, T. et al., Clin Cancer Res 6: 3530-5 ( 2000), Muller, A. and collaborators Nature 410: 50-6 (2001), Robledo et al, J Biol. Chem 276: 45098-45105 (2001)) and in an example link of this expression with cell metastasis of breast tumor (Muller, A. et al, Nature 410: 50-6 (2001).) To further investigate the function of chemokine receptors on tumor cells, the inventors carried out the evaluation of CSCR4 expression in several lines of human breast tumor cells Initially the expression pattern of CXCR4 was evaluated . by flow cytometry T lymphocytes cultured with primary IL-2 and two T-cell lines, CEM-NKr and Jurkat, were examined to determine the T-cell phenotype of anti-CXCR4 spotting. Three lines of breast tumor cells, MCF-7, MDA MB-231 and MDA MB-435s, were also tested. All or the four tested anti-CXCR4 clones stained T cells. Surprisingly, while breast tumor cells are reported to express CXCR4, the widely used 12G5 clone did not detect any CXCR4 on breast tumor cells. The weak and variable reactivity s.e 5 detected with the other three tested clones on the breast tumor cells. The DU 4475 and ZR 75-1 tumor cell lines were also tested in this assay (data not shown) and found to have antibody staining profiles similar to the other 0 breast tumor cells tested. Thus, mAb panel staining patterns for CSCR4 are considered to suggest two distinct types of reactivity: a "leukocyte" CXCR4 phenotype (exemplified by CEM-NKr, Jurkat and IL-2 lymphocyte staining) and a cell phenotype of breast tumor (exemplified by weak spotting on the breast tumor cell line MCF-7 and MDA MB-231). The consistent lack of reactivity using the clone 12G5 anti CXCR4 mAb widely used, on breast tumor cells led the inventors to examine the expression of CXCR4 in these cells by RT PCR. The mRNA was isolated from the three breast tumor lines tested in flow cytometry as well as the lymphocytes cultured with IL-2 and the T cell lines, CEM-NKr and Jurkat, as positive controls for CXCR4 expression. Despite the lack of reactivity with 12G5 and the variable reactivity with the other anti-CXCR4 clones tested, the breast tumor cell lines, MCF-7 and MDA MB-231, expressed the message of CXCR4; however, MDA MB-435s was found to be negative for CXCR4 expression. All cases GAPDH was measured as a control. To examine whether differences in mAb reactivity could be due to sequence differences thus resulting in epitope variations in CXCR4 over several cell lines, the inventors then sequenced the PCR products generated from MCF-7, as a representative CXCR4 + breast tumor cell and CEM-NKr, as a representative T cell. The sequences of these two cell lines are identical to the published CXCR4 sequence suggesting that despite the different CXCR4 antibody profiles, the genetics and thus the structure of CXCR4 polypeptide in both cell types was identical. The inventors have previously reported a set of techniques whereby receptor binding to a comprehensive arrangement of chemokine ligands can be simultaneously estimated (Dairaghi et al J. Biol. Chem 274: 21569-74 (1999); Gosling J et al. J Immunol 164: 2851-6 (2000)). In this way the inventors probed the CXCR4 linkage profile in CEM-NKr as compared to the MCF-7 cells. Greater than 90 chemokine elements were tested for the ability to displace the chemokine signal, 125I SDF-la, for the CEM-NKr link (Figure 1) or MCF-7 cells (Figure 1). As expected, the potential high affinity competitors of 125I SDF-la in CEM-NKr include hSDF-lß and mSDF-1, while hSDF-la and HHV8 vMIP-II exhibit potential moderate affinity competition. This is consistent with all the previously reported results of SDF-1 as the only non-viral ligand of CXCR4. However, the overall pattern of competition in MCF-7 cells was markedly different. In this type of cell, l-TAC and ml-TAC demonstrated high affinity competition for the same SDF-1 signal ligand. To further investigate unusual result 125I I-TAC was tested as the signal ligand in MCF-7 cells (Figure 1). The high affinity displacement profile using 125 I I-TAC on MCF-7 was identical to the profile obtained using 125 I SDF-la. Thus, in MCF-7 cells I-TAC and SDF-1 behave indistinguishably in the binding and compete for the same receptor site. To further characterize the I-TAC and SDF-1 binding, dose response curves were obtained in competition binding experiments with potential high affinity ligands selected on CEM-NKr and MCF-7. As suggested by DisplaceMax ™ data, I-TAC competes with 125I and SDF-la for binding to MCF-7, but not CEM-NKr (Figure 2). The 125ISDF-la homologous competition with either the isoform SDF-1, SDF-la or SDF-lβ, resulted in complete competition over CEM-NKr and MCF-7 (Figure 2). Notably, the affinity of SDF-1 for the receptor expressed in MCF-7 is higher than that in CEM-NKr. Thus, while the sequence of CXCR4 is identical in both cell types the binding specificity of ligand in affinity differ in T cells against breast tumor cells. The investigators then investigated whether the I-TAC binding detected in MCF-7 cells could be mediated by CXCR3 since CXCR3 has long been established as the main receptor for I-TAC (Cole, KE et al., J Exp Med 187 : 2009-21. (1998)). To this end, the 125 I I-TAC link was examined under conditions that would exhibit the linkage mediated by CXCR3 Ylasic '(ie the binding of the reported CXCR3 ligands MIG, I-TAC and IP-10 to CXCR3) allowing this linkage mediated by CXCR4 Y '(that is, the linkage of the ligand CXCR4 reported SDF-1 to CXCR4) as well as the inverse situation. MCF-7 cells were pre-incubated with either the medium alone, medium containing MIG (~ 175 nM, to inhibit CXCR3-mediated binding) or medium containing excess SDF-lβ (~90 nM, to inhibit mediated binding) by CXCR4, I-TAC competed with 1? 5I I-TAC for binding to MCF-7 cells or an IC50 of InM (Figure 3) confirming that it is a high affinity ligand for this receptor on these cells., cells first preincubated with MIG in excess were then able to give the same binding curve of I-TAC 125I I-TAC homologous again with an IC50 of 1 nM (Figure 3). However, when the cells were first pretreated with SDF-lβ in excess the binding of 125 I I-TAC was inhibited (Figure 3) suggesting that the 125 I I-TAC link observed on the breast tumor cells is mediated by the receptor SDF1 expressed on these cells. Similarly, the binding of 125 I-TAC to MCF-7 cells was not inhibited when IP-10 was tested as the cold chemokine competitor. Again the preincubation with MIG in excess did not make this link profile; however preincubation of the cells with SDF-lβ completely inhibited the binding of lz5I I-TAC. When the ligand of CXCR3 MIG was tested as the binding of 125 I I-TAC of cold competitor to the cell was not inhibited (Figure 3). As expected from the DisplaceMax ™ data depicted in Figure 1, SDF-lβ competed with 1 5 I I-TAC for the binding of these cells with high affinity (IC50 of 1 nM). Preincubation of cells with excess MIG did not compete for SDF-lß / 125I I-TAC, again suggesting the detected link that is not mediated by CXCR3. This hypothesis was further examined by PCR. Isolated mRNA previously used (described above) was used to probe the evidence of CXCR3 transcripts. Whereas, lymphocytes cultured with IL-2 expressed CXCR3; no other cell tested expressed CXCR3. The lack of expression of CXCR3 detected by RT PCR supports the data of Figure 3, again suggesting that the I-TAC binding on MCF-7 cells is not mediated by CXCR3. The reactivity of altered anti-CXCR4 antibody as well as the altered ligand binding affinity specificity led the inventors to consider that this receptor was not classical CXCR4. There are a few 'orphan chemokine' receptors that have been identified, but for which the chemokine ligand has not been identified. The inventors considered several orphan receptors. A receiver of such kind is called RDC1 (referred to herein as CCX-CKR2). When the protein sequence for RDC1 is transfected into MDA MB 435s (a cell line that does not endogenously express CSCR4, CXCR3 or CCX-CKR2) the labeled radioligand binding phenotype is recapitulated (Figure 4). CCX-CKR2 expressed in MDA MB 435s binds to radiolabelled SDF-1. This link is competed by cold competitors SDF-1 and I-TAC. In the effort to target this receptor with small molecular weight organic compound (SMC) therapeutics, the inventors classified small molecules (nearly 135,000) using two high-throughput classifications: one designed to estimate the SDF-1 linkage phenotype of leukocyte mediated by CXCR4 and one to probe the phenotype of SDF-1 linkage of breast cancer mediated by CCX-CKR2. The results of these classifications indicated that clear pharmacological discrimination of the two linkage phenotypes was possible (Figure 5). For example, the small molecule designated CCX0803 competes with 125I SDF-la for the MCF-7 binding with an IC50 of 46 nM (Figure 5), however, this small molecule does not inhibit the binding of 125I SDF-la in CEM-NKr in everything (Figure 5). In contrast, a "different" small molecule antagonist, CCS7923, inhibits the binding of 1"5I SDF-la to CEM-NKr an IC50 of 106 nM (Figure 5), but does not inhibit the binding of 125I SDF-la on cells MCF-7 (Figure 5). These two compounds reveal the marked and unambiguous pattern of the inhibition of non-reciprocal binding of ligand to the two receptors (breast tumor lines against leukocytes). After initially determining that the breast cancer cells exhibit a binding affinity for SDF-1 which is different from that observed on other non-tumor or non-cancerous tissue, additional studies were performed. These phenotype studies (using antibody reactivity, ligand binding profile and pharmacological discrimination, see detailed methods herein), have clearly shown that many types of cancer (or tumor) cells also exhibit the affinity of linkage (eg, antibody reactivity, ligand binding, and pharmacological discrimination) initially correlated with breast tumor cells and thus the expression of CCX-CKR2. The following tumor cells were examined and exhibited the correlation affinity correlated with cancer: human ovarian carcinoma, human cervical adenocarcinoma, human Burkitt's lymphoma, human breast adenocarcinoma, human breast ductal carcinoma, human glioblastoma and mouse mammary tumor. Tumors and other cancers are difficult to treat in part because of their rapid rate of cell growth. In this aspect, tumors are known to share some growth characteristics with rapidly dividing early embryonic tissues. A teaching school suggests that tumors in the adult may represent Aevertors' to an embryonic growth phenotype. Both of the genetically inactivated mice SDF-1 and CXCR4 are lethal embryos, suggesting that this pair of ligand receptor is a critical component of growth growth. Approximately 50% of homozygous mutant SDF-1 embryos died perinatally by 18.5; the remaining homozygous baits died within 1 h of birth (Kishimoto, et al., Nature 382: 635-638 (nineteen ninety six) ) . Similarly, ~ l / 3 of the inactivated mice CXCR4 homozygotes died perinatally at E18.5 (Ma, et al, Proc Nati, Acad Sci USA 95: 9448-9453 (1998). In both of the inactivation defects of receptor and ligand defects of lymphopoiesis and myelopoiesis were observed. The fetal liver is the largest site of hematopoiesis in the mouse on day 11 and continues as such until the first post-natal week. For this purpose the inventors decided to examine the expression of CCX-CKR2 in this compartment. The inventors examined the expression CCX-CKR2 in E17 wild-type mouse embryos (a point in development close to the time the inactivated animals die) E13 (a developmental point other than the time the inactivated animals died, still after that hematopoiesis begins).
In SDF-1 binding assays, radiolabeled human SDF-1 binds E13 fetal liver cells and both SDF and I-TAC (mouse and human proteins) are able to compete with the radiolabeled tracer for binding. This altered ligand specificity as exemplified by I-TAC binding to the SDF-1 receptor is a hallmark of the binding phenotype that the inventors first correlated with cancer cells and have now shown that CCX-CKR2. In addition, CCX-CKR2 antagonists are able to compete with the SDF-1 link in the fetal E13 liver; however, CXCR4 antagonists do not compete. Later in development, the fetal liver cells in E17 express CXCR4 and these cells respond to SDF-1 by mobilizing intracellular calcium. CXCR4 antagonists inhibit this calcium mobilization mediated by SDF-1 however, the CCX-CKR2 antagonists have no effect. Thus, these data suggest that wild-type fetal liver cells in E13 and E17 both express CXCR4, however, CCX-CKR2 is expressed early (Eli) but not at later time points (E15). Although linkage studies in embryonic mouse models correlate well with data from human studies, preliminary experiments using mice that have a targeted disruption of the CKR4 gene suggest that the SDF-1 and I-TAC binding profiles observed in the Embryonic day 13 (E13) of the fetal liver cells are unchanged. This provides additional evidence that the gene encoding the polypeptide with the cancer-related SDF-1 binding affinity is not CXCR4. The experiments also demonstrate that the CCX-CKR2 receptor can provide a stimulatory signal for the growth of tumor cells. Tumor cells can upregulate certain genes involved in the cell cycle or transcription in response to stimulation of SDF-1. More importantly, if the tumor cells are removed from the serum in the culture overnight and they begin to go through apoptosis (cell death). When SDF-1 is added to supplement these crops the cells are able to recover from starvation, as compared to untreated controls. Thus SDF-1 therefore serves as an antiapoptotic signal. Cancer cells are often characterized as cells that have lost the ability to undergo apoptosis. 20. Example 2: This example demonstrates that the cancer-related binding phenotype discussed in Example 1 is mediated by CCX-CKR2 (previously known as the orphan receptor RDC1). In general, CCS-CKR2 is preferentially expressed in transformed cells. As shown in Table 1 (left column), a variety of different cancer cells were tested positive for the expression of CCX-CKR2.
In contrast, more normal (non-tumor) cells did not express CCX-CKR2. See, Table 1 (right column). Table 1 CCX-CKR2 positive CCX-CKR2 negative Human Mammary Carcinoma (MCF-normal human PBMC 7, MDA MB 361) Human glioblastoma (T98G) Human T-cell leukemia (M0LT4, Jurkat, CEM-NKr) Human Prostate Carcinoma Endothelial cells do not (LN Cap) stimulated Human B cell lymphoma Mouse thymus (Raji, IM9) Human Ovarian carcinoma Mouse lung (HeLa) Human lung carcinoma Mouse spleen (A549) Carcinoma Mouse mammary Mouse heart (4T1) PBL epithelial cells mouse pancreatic, SV40 transformed (SVR) Mouse B cell lymph Mouse liver (BCL1) Mouse normal kidney * Mouse adult total bone marrow Normal mouse Negative adult bone marrow Brain * Mouse lineage Fetal mouse liver (Eli Fetal mouse liver (E15 to E13) until birth) Activated endothelial cells * The expression in these organs is weak as determined by the radioligand binding signal However, it is presented which is a function for CCX-CKR2 in some normal cells. The CCX-CKR2 receptor is expressed for a period of time in fetal development. CCX-CKR2 then in the mouse fetal liver by the 11th day of embryogenesis (Eli), but by E15 it is not detected any longer (as determined by the radiolabelled SDFl binding and the I-TAC shift) as well as the CCX-CKR2 transcripts detected by the Northern analysis. In the adult mouse this is expressed in normal kidney. By comparison in kidney expression, there is a lower expression in the normal brain. Because the test becomes a complete brain homogenate this low signal in the radioligand binding assay is consistent with a small population of cells in the brain that express CCX-CKR2. To further reinforce the evidence that the function of CCX-CKR2 in cancer, the inventors demonstrated that the growth of the cancer cell can be inhibited by antagonizing CCX-CKR2 in cancer cells. Antagonism of CCX-CKR2 expressed in a mammary carcinoma by a CCX-CKR2 antagonist inhibited cell proliferation in vitro. Cells treated in vitro exhibited reduced cell growth over time as compared to untreated controls. See, Figure 6. CCX-CKR2 is also involved in adhesion. The migration of leukocytes involves several stages that include the addition of cells and subsequent immigration to a given tissue. Static adhesion in vitro analyzes the model of this event. The monolayers of tilted endothelial cells are grown on a surface. The cells expressing CCX-CKR2 are then labeled with a fluorescent dye for visualization. When the CCX-CKR2 cells are allowed to adhere to the endothelial surface much more cells expressing CCX-CKR2 bind to the endothelial layer than the control cells of CCX-CKR2 do. In addition, the addition of a CCX-CKR2 antagonist inhibits adhesion as compared to a vehicle treated control. See, Figure 7. The in vivo evidence further supports a function for CCX-CKR2 in tumor growth. Tumors are formed when human B cell lymphoma cells, which express CCX-CKR2, are injected into immunodeficient mice. Treatment of these mice with CCX-CKR2 antagonist inhibited vascularized tumor formation. In one such study, one of 17 mice treated with a CCX-CKR2 antagonist developed a vascularized, encapsulated tumor while 11 of 17 mice in the vehicle-treated group developed encapsulated vascularized tumors. These data suggest that CCX-CKR2 may be involved in a tumor's ability to differentiate and establish a vascular bed and provides evidence that antagonism of CCX-CKR2 is a useful cancer therapy. The effect of CCX-CKR2 antagonism was also tested in a breast cancer model. In a breast tumor growth model, immunodeficient mice were injected with a human mammary carcinoma. Tumor measurements were made 3 times a week and volumes were plotted. Mice that were treated with a CCX-CKR2 antagonist exhibited reduced tumor volume as compared to the vehicle control group, which demonstrates that CCX-CKR2 has a role in tumor growth. See, Figure 8. Example 3 This example demonstrates that CCX-CKR2 promotes cell survival by reducing apoptosis.
Interactions between chemokines and chemokine receptors are typically estimated by measuring intracellular calcium mobilization and chemotaxis. However, CCX-CKR2 did not produce a transient calcium mobilization or cause the cells to migrate in response to their CXCL12 or CXCL11 ligands. Cells expressing CCX-CKR2 however exhibit increased addition to activated endothelial cell monolayers. In addition, under conditions of low serum culture medium supplementation (ie, 1% instead of 10% regular), the recovery of live adherent cells after three days was much greater for transfectants CCX-CKR2-MDA MB 435s (designated CCX-CKR2 435s) against non-transfected WT cells (WT 435s). Consistent with this observation, the frequency of dead cells recovered in the supernatant collected from these cultures was much larger for WT against the CCX-CKR2 transfectants. This effect could be visualized fluorescently using the intercalation dye of DNA 7AAD (7 aminoactinomycin D). The transfectants CCX-CKR2-435s or. Wild type 435s cells were cultured in different serum concentrations, then harvested and incubated with 7AAD (1 ug / ml in DMSO-) for 15-30 minutes at room temperature. The FACS analysis revealed much more dead / apoptotic cells (i.e., 77? AD-positive) in wild-type 435s cells against the CCX-CKR2-435s transfectants. The inventors have now extended these findings in a series of experiments where cultured CCX-CKR2 transfectants or non-transfected WT cells were co-stained with Annexin and detects only apoptotic cells and propidium iodide (PI) that detects dead cells but not apoptotic cells . This method readily identifies the proportion of apoptotic cells in a population of cells, as demonstrated using known agents to induce cellular apoptosis, for example, camptothecin (CMP), or TNFalpha cycloheximide (CHX) and which provide excellent controls in these assays. Using this assay, the inventors measured the development of apoptotic cells over time of CCX-CKR2-435s transfectants or cultured wild type 435s cells either in optimal (10%) or limiting (1%) serum. Both types of cells cultured in 10% serum showed excellent viability over a culture period of 4 days. In contrast, WT cells cultured in 1% serum showed a dramatic reduction in viable cells after 3 and 4 days of culture. Co-staining with Annexin and Pl revealed this reflected development of both apoptotic and dead cells. Interestingly, CCX-CKR2-435 cells cultured in 1% serum showed excellent viability over the same 4-day culture period, suggesting that the introduction of CCX-CKR2 into these 435s protected cells from rapid cell apoptosis occurs under sub-optimal serum supplementation conditions. Identical results were obtained in a second experiment using the same CCX-CKR2-435s transfectant, or in addition to a non-clonal population separated from 435s cells transfected with CCX-CKR2. These latter results indicated that the poor ownership of apoptosis of the initial clonal transfectant resulted from the CCX-CKR2 expression before a particular aberration of a transfectant clone. Example 4 This example demonstrates phosphorylation mediated by CCX-CKR2 from p44 / 42 MAPK (ERK1 and ERK2). CCX-CR2 is an unusual ligand binding receptor (for example, by ITAC or SDF1) does not result in the mobilization of calcium that is typical of many chemokine receptors. This hastened an assessment of other potential signaling pathways, including an investigation into whether the activity of CCX-CR2 was mediated through ERK phosphorylation. Experiments conducted with lysates of an MCF7 cell line expressing CCX-CR2 that was stimulated with either ITAC or SDF1 demonstrated that there is a ligand-dependent phosphorylation of ERK1 and ERK2 (sometimes also referred to in the literature as p44 / 42 MAPK) . Phosphorylated ERK1 and ERK2 were detected by Western blot analysis, with lysates of the MCF7 cell lines initially subjected to electrophoresis to separate proteins in the lysate. Phosphorylated ERK1 and ER2 on the electrophoretic gel were detected by probing with specific antibodies to the phosphorylated form of these proteins. These antibodies are available from Cell Signaling Technologies of Beverly, MA. Similar experiments were conducted with Hela cells expressing CCX-CR2. The same results were obtained when these cells were stimulated with either ITAC or SDF1. Since ITAC and SDF1 bind to other chemokine receptors, eg, CXCR3 in the case of ITAC, and CXCR4 in the case of SDF1, it was important to consider the potential contribution of these receptors to the ERK phosphorylation induced by these ligands in cells MCF7. In fACS analysis of MCF7 cells using antibodies specific for CSCR3 or CXCR4 revealed a complete absence of these receptors in MCF7 cells. Positive controls using cell lines that express CXCR3 or CXCR4 validated that the antibodies used in these experiments could bind these receptors. These data in this manner indicate that binding ligands such as ITAC or SDF1 to CCX-CR2 cause intracellular signaling via an ERK phosphorylation, which likely results in the activation of additional downstream components. The discovery that CCX-CR2 mediates phosphorylation of ERK1 and ERK2 indicates that CCX-CR2 is associated with a variety of biological processes because the phosphorylation of ERK has been shown to mediate the regulation of cell growth and cell differentiation. Example 5 This example demonstrates that the cellular expression of CCX-CKR2 causes the induction of numerous regulatory proteins. As an alternative procedure to investigate the signaling events mediated by CCX-CKR2, the supernatants harvested from the MDA MB 435s cells transfected with CCX-CKR2 were compared to the supernatants harvested from the MDA MB 435s (435s) wild type cell, evaluated by specific ELISA assays for the presence of a large family of secreted proteins. 435s cells expressing CCX-CKR2 produced substantially larger amounts of GM-CSF, RANTES, MCP-1, TIMP-1 and MMP3 than wild-type 435s cells, especially when cultured under limiting serum conditions. Interestingly, all of these factors have been reported to be involved in growth, vascular remodeling and chemotaxis related to tumorigenesis. They may also be involved in the scarce phenotype of apoptosis of CCX-CKR2 described in the above. Example 6 This example demonstrates siRNA-based inhibition of CCX-CKR2. The inventors obtained specific SMARTpool ™ siRNA (Dharmacon) for either CXCR4 or CCX-CKR2. The SMARTpóol ™ siRNA is an accumulation of four different siRNA sequences. Each one with direction to a different region of the specific mRNA. These accumulations of siRNA were tested in HeLa cells. The CXCR4 expression was estimated by staining 12G5 or 173 Mab and FACS, while the expression of CCX-CKR2 was measured in a binding assay using 125I-SDF1. CXCR4 is expressed on HeLa cells in a conformation that does not exhibit detectable 125I-SDF1 binding, thus allowing the detection of CCX-CKR2 expression. siRNA SMARTpoll ™ CCX-CKR2 (25-100 nm) effects significant inhibition (>50%) of the 125I-SDF1 link, whereas siRNA SMARTpool ™ CXCR4 did not. Similar results were obtained with the transfectants 293-CCX-CKR2. In addition, the following 3 siRNA sequences were each found to reduce the SDF-1 binding when introduced into cells at a concentration as low as 4 nM: siRNA # 1: GCCGTTCCCTTCTCCATTATT siRNA # 2 GAGCTCACGTGCAAAGTCATT siRNA # 3: GACATCAGCTGGCCATGCATT Example 7 This example demonstrates the expression of CCX-CKR2 in the brain. CCX-CKR2 is expressed on a large number of tumor cell lines, but a few normal tissues. An exception to this latter pattern was provided by the demonstration in the binding studies of the brain cells of normal adult mice expressed CCX-CKR2. To extend these observations, in situ hybridization studies were performed using a specific CCX-CKR2 probe to locate the region of CCX-CKR2 expression within the brain. Brain samples were collected from normal adult mice and fixed with 4% PFA in PBS overnight at 4 ° C, followed by 30% sucrose in PBS overnight at 4 ° C. The tissues were then embedded in OCT, cut into 20 slices of um, then harvested on the super-frozen plates. For the in situ hybridization studies, the antisense and sense riboprobes were prepared by in vitro transcription, respectively, with T7 and SP6 RNA polymerase using the DIG cRNA (Roche) labeling kit after the linearization. The 20 μm cryosections were fixed in fresh 4% PFA, followed by K protease treatment (2 ug / ml for 20 min at 37 ° C). The platens are prehybridized at 55 ° C for 1 hour, and then hybridized at 55 ° C O / N in sealed containers. The platens are then washed with 50% formamide, 5x SSC pH 4.5 and 1% SDS at 65 ° C followed by blocking with 5% sheep serum for one hour and incubation with 1: 1000 anti-DIG antibody. in 1% sheep serum for O / N at 4 ° C. The platens were washed with TBST and detected with NBT / BCIP. The results clearly demonstrate the strong expression of CCX-CKR2 by neurons within the cerebellum, hippocampus and cortex. There is little or no detectable expression in areas containing equal cells such as the tracts of white matter in the cerebellum, and the corpus callosum. Purkinje cells showed strongly positive signal evenly in subset of the granule cells in the inner granule cell layer showed positive signal. In addition, the bark is generally very positive, and no individual inside the bark showed clear positive staining. The hippocampus showed strong signal of CCX-CKR2 in the neurons of the dentate gyrus and CA1-3. In addition, the superimposed crusts were also positive. These data provide some insight that considers the potential relevance of CCX-CKR2 to brain tumors. CCX-CKR2 is not expressed in cells of the ventricular zone, or the gual cells, where astrocytomas are thought to originate. In contrast, there is some expression in what is presented to be a subset of cerebellar granule cells, the type of cell from which medulloblastoma is derived. The expression profile for CCX-CKR2 is very different from that observed for the other SDF-1 CSCR4 receptor as described by others. Example 8 This example demonstrates the efficacy of a ligand competitor of CCX-CKR2 in a mouse half-graft model of lung carcinoma. Lung carcinoma is the leading cause of death from cancer in the United States. CCX-CKR2 is expressed in lung carcinoma as well as in activated endothelium. The effects of administering a CCX-CKR2 ligand competitor, a compound of the 700 series, in a lung carcinoma semi-graft model was evaluated. The semi-graft study of lung carcinoma, A549 tumor fragments (30-40 mg) were implanted in the subcutaneous space in nude mice. Tumors were allowed to develop up to approximately 150 mg in size (between 100 and 200 mg) in which mice were enrolled in the study and treatment was started. Mice were treated with the ligand competitor CCX-CKR2 (25 mpk; administration, QID) or vehicle control. Melphalan was included as the positive control (9 mpk / dose, ip administration, Q4Dx3). Tumors were measured twice a week with a calibrator in two dimensions and converted to tumor mass using the formula for a prolate ellipsoid (ax b2 / 2), where a is the longest dimension and b is the shortest dimension, and assuming the unit density (1 mm3- 1 mg). Body weights were also measured twice a week to estimate any of the adverse effects of the compound dosage. The antitumor activity was estimated by the growth retardation of the tumor of the treated group compared to the control group treated by vehicle. The mice receiving the competitor exhibited reduced tumor load compared to the group treated per vehicle. This difference in tumor volume was statistically significant between these groups and represents a 32% reduction in the average tumor volume. Melphalan also reduced tumor volume with a 60% reduction in the average tumor volume. Additionally, daily treatment with the competitor was well tolerated in this study as determined by the weight gain in the animals treated with the compound consistent with that of the vehicle-treated mice.
The tumor weights were estimated on the final day of treatment with the compound (day 49). Mice treated with the CCX-CKR2 competitor exhibited tumors that were statistically smaller than those in the vehicle control group. Similarly, mice receiving Melphalan also had significantly smaller tumors than the vehicle-treated group. All publications and patent applications cited in this specification are incorporated herein by reference as if each individual publication or patent application was specifically and individually indicated to be incorporated by reference. Although the above invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it will be readily apparent to one of ordinary skill in the art in view of the teachings of this invention, that certain changes and modifications can be made to it without departing from the spirit and scope of the appended claims.

Claims (45)

  1. CLAIMS 1. A method for identifying an agent that binds CCX-CKR2 on a cell, the method characterized in that it comprises, - contacting a plurality of agents to a CCX-CKR2 polypeptide comprising at least 95% identical extracellular domain to an extracellular domain of SEQ ID NO: 2, or a fragment thereof that binds SDF1 or I-TAC; and selecting an agent that competes with I-TAC or SDF1 for binding to the CCX-CKR2 polypeptide or fragment thereof, in order to thereby identify an agent that binds CCX-CKR2 on a cell.
  2. 2. The method according to claim 1, characterized in that the cell is a cancer cell.
  3. 3. The method of compliance with the claim 1, characterized in that it further comprises testing the agent selected for the ability to bind to, or inhibit the growth of, a cell.
  4. 4. The method according to claim 3, characterized in that the cell is a cancer cell.
  5. 5. The method according to claim 1, characterized in that it further comprises testing the agent selected for the ability to alter the function of the kidney.
  6. 6. The method according to claim 1, characterized in that it further comprises testing the agent selected for the ability to alter brain or neuronal function.
  7. 7. The method according to claim 1, characterized in that it further comprises testing the agent selected for the ability to change cell damage to endothelial cells.
  8. 8. The method according to claim 1, characterized in that the agent is less than 1,500 daltons.
  9. 9. The method according to claim 1, characterized in that the agent is an antibody.
  10. 10. The method according to claim 1, characterized in that the agent is a polypeptide.
  11. 11. The method according to the claim 1, characterized in that the CCX-CKR2 polypeptide comprises the sequence shown in SEQ ID NO: 2.
  12. 12. A method for determining the presence or absence of a cancer cell, the method characterized in that it comprises contacting a sample comprising a cell with an agent that specifically binds to SEQ ID NO: 2; and detecting the binding of the agent to a polypeptide in the sample, wherein the binding of the agent to the sample indicates the presence of a cancer cell.
  13. 13. The method according to claim 12, characterized in that the agent is an antibody.
  14. 14. The method according to claim 12, characterized in that the agent is less than 1500 daltons.
  15. 15. The method according to claim 12, characterized in that the agent is a polypeptide.
  16. 16. The method according to claim 12, characterized in that the polypeptide detected is the SEQ ID NO: 2.
  17. The method according to claim 12, characterized in that the sample is from a human.
  18. 18. The method according to claim 12, characterized in that the method is used to diagnose cancer in a human.
  19. 19. The method according to claim 12, characterized in that the method is used to provide a prognosis of cancer in a human.
  20. 20. The method of compliance with the claim 12, characterized in that the cancer is selected from the group consisting of cervical cancer, breast cancer, lymphoma, glioblastomas, prostate cancer and leukemia.
  21. 21. The method according to claim 12, characterized in that the cancer is not Kaposi sarcoma, multicentric Castleman's disease or primary effusion lymphoma associated with AIDS.
  22. 22. The method according to claim 12, characterized in that the antibody competes with SDF1 and I-TAC for binding to SEQ ID NO: 2.
  23. 23. A method for providing a diagnosis or prognosis of an individual having cancer, the method characterized in that it comprises detecting the presence or absence of expression of a polynucleotide encoding a CCX-CKR2 polypeptide in a cell of an individual, wherein the CCX-CKR2 polypeptide binds to I-TAC and / or SDF1 and the CCX-CKR2 polypeptide it is at least 95% identical to SEQ ID NO: 2, in order to diagnose a cancer in an individual.
  24. 24. The method of compliance with the claim 23, characterized in that the CCX-CKR2 polypeptide is shown in SEQ ID NO: 2.
  25. 25. The method according to claim 23, characterized in that the cancer is selected from the group consisting of cervical cancer, breast cancer, lymphoma, glioblastomas, prostate cancer, and leukemia.
  26. 26. The method according to claim 23, characterized in that the cancer is not sarcoma de. Kaposi, multicentric Castleman's disease or primary effusion lymphoma associated with AIDS.
  27. 27. An antibody, characterized in that it specifically competes with SDF-1 and I-TAC for the binding to SEQ ID NO: 2.
  28. 28. The antibody according to claim 27, characterized in that the antibody is a monoclonal antibody.
  29. 29. The antibody according to claim 27, characterized in that the antibody is a humanized antibody.
  30. 30. A method, characterized in that it comprises, contacting a cell with an agent that specifically binds to SEQ ID NO: 2, wherein the agent competes with SDF-1 and I-TAC for binding to a CCX-CKR2 polypeptide, wherein the cell expresses a CCX-CKR2 polypeptide comprising an extracellular domain at least 95% identical to an extracellular domain of SEQ ID NO: 2, in order to bind the agent to the CCX-CKR2 polypeptide on the cell.
  31. 31. The method according to claim 30, characterized in that the agent is less than 1,500 daltons.
  32. 32. The method according to claim 30, characterized in that the agent is an antibody.
  33. 33. The method according to claim 30, characterized in that the agent is a polypeptide.
  34. 34. The method according to claim 30, characterized in that the CCX-CKR2 polypeptide is as shown in SEQ ID NO: 2.
  35. 35. The method according to claim 30, characterized in that the agent is identified by a method comprising contacting a plurality of agents to a CCX-CKR2 polypeptide comprises at least an 95% extracellular domain identical to an extracellular domain of SEQ ID NO: 2, or a fragment thereof that binds SDF1 or I-TAC; and selecting an agent that competes with I-TAC or SDF-1 for binding to the CCX-CKR2 polypeptide or fragment thereof, in order to thereby identify an agent that binds a cancer cell.
  36. 36. A method for treating cancer in an individual, the method characterized in that it comprises administering to the individual a therapeutically effective amount of a polynucleotide that inhibits the expression of a polynucleotide CCX-CKR2.
  37. 37. The method according to the claim 36, characterized in that the polynucleotide CCX-CKR2 encodes SEQ ID NO: 2.
  38. 38. The method according to the claim 36, characterized in that the polynucleotide CCX-CKR2 comprises SEQ ID NO: l.
  39. 39. A method for treating cancer in an individual, the method characterized in that it comprises administering to the individual a therapeutically effective amount of an agent competing with SDF1 and I-TAC for binding to SEQ ID NO: 2.
  40. 40. The method according to claim 39, characterized in that the agent is less than 1,500 daltons.
  41. 41. The method according to claim 39, characterized in that the agent is an antibody.
  42. 42. The method of compliance with the claim 39, characterized in that the agent is a polypeptide.
  43. 43. The method according to claim 39, characterized in that the agent is identified by a method comprising contacting a plurality of agents with a CCX-CKR2 polypeptide comprising an extracellular domain at least 95% identical to an extracellular domain. of the SEQ ID NO: 2, or a fragment thereof that links SDFl or I-TAC; and selecting an agent that competes with I-TAC or SDF-1 for binding to the CCX-CKR2 polypeptide or a fragment thereof, to thereby identify an agent that binds to a cancer cell.
  44. 44. The method according to claim 39, characterized in that the cancer is selected from the group consisting of cervical cancer, breast cancer, lymphoma, glioblastomas, prostate cancer and leukemia.
  45. 45. The method according to claim 39, characterized in that the cancer is not Kaposi's sarcoma, multicentric Castleman's disease or primary effusion lymphoma associated with AIDS.
MXPA/A/2006/004843A 2003-10-30 2006-04-28 Compositions and methods for detecting and treating diseases and conditions related to chemokine receptors MXPA06004843A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US10698541 2003-10-30
US10912638 2004-08-04

Publications (1)

Publication Number Publication Date
MXPA06004843A true MXPA06004843A (en) 2006-12-13

Family

ID=

Similar Documents

Publication Publication Date Title
US20110165164A1 (en) Compositions and methods for detecting and treating diseases and conditions related to chemokine receptors
IL175179A (en) Method of screening for an agent that binds to ccx-ckr2 on a cell and its use in detecting the presence or absence of a cancer cell
KR20060136373A (en) Compositions and methods for detecting and treating diseases and conditions related to chemokine receptors
KR100961237B1 (en) Compositions and methods for detecting and treating diseases and conditions related to chemokine receptors
Hoffmann et al. Rapid uptake and degradation of CXCL12 depend on CXCR7 carboxyl-terminal serine/threonine residues
Dauphinee et al. SASH1 is a scaffold molecule in endothelial TLR4 signaling
Montuori et al. The cross-talk between the urokinase receptor and fMLP receptors regulates the activity of the CXCR4 chemokine receptor
JP4916889B2 (en) Methods and compositions for modulating angiogenesis
US7413866B2 (en) Compositions and methods for detecting and treating diseases and conditions related to chemokine receptors
Kollis et al. Characterising distinct migratory profiles of infiltrating T-cell subsets in human glioblastoma
US20110052667A1 (en) Compositions and methods for inhibiting angiogenesis and tumorigenesis
US20040259152A1 (en) Novel methods of diagnosing and screening for modulators of tissue remodeling and treating related diseases
MXPA06004843A (en) Compositions and methods for detecting and treating diseases and conditions related to chemokine receptors
Denisov et al. 23P Transcriptional profiling of circulating tumor and hybrid cells in breast cancer patients
Miranda et al. N-MYC impairs innate immune signaling in high-grade serous ovarian carcinoma
US20100247540A1 (en) Methods and Compositions For Modulating Angiogenesis
KR101149115B1 (en) Methods and compositions for modulating angiogenesis
US20180348216A1 (en) Cd318 as a marker for, and cd318 inhibition as a treatment for, autoimmune disease
O'Hayre Elucidating the functions and signaling mechanisms of the chemokine CXCL12 and its receptors CXCR4 and CXCR7 in cancer
Cain Development of surface proteome signature technology and identification of CD44 as an anti-apoptotic mediator